METHODS FOR TREATING HYPOPHOSPHATASIA IN CHILDREN

The disclosure features methods for treating hypophosphatasia (HPP) in a patient (e.g., a child having HPP) exhibiting physical impairments, disability in activities of daily living (ADL), pain, and/or delayed motor development by administering a soluble alkaline phosphatase (sALP) to the patient.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
FIELD

The disclosure relates to methods for treating hypophosphatasia (HPP).

BACKGROUND

Hypophosphatasia (HPP) is a rare, heritable skeletal disease with an incidence of 1 per 100,000 births for the most severe forms of the disease. HPP is often fatal when observed at birth, having an infant mortality rate of ˜70%. Severely affected patients often die in infancy from respiratory insufficiency due to progressive chest deformity.

The disorder results from loss-of-function mutations in the gene coding for tissue-nonspecific alkaline phosphatase (TNALP). HPP leads to a remarkable range of symptoms and severity, from rickets (osteomalacia) to almost complete absence of bone mineralization in utero. Most patients exhibit the characteristics of skeletal changes, short stature, painful lower limbs, gait disturbance, and premature shedding of teeth. For instance, infantile symptoms of HPP can include inadequate weight gain, the appearance of rickets, impaired skeletal mineralization, progressive skeletal demineralization, rib fractures, and chest deformity, while childhood symptoms of HPP can include short stature and skeletal deformities, such as bowed legs and enlarged wrists, knees, and ankles as a result of flared metaphyses. Due to physical impairments associated with HPP, patients afflicted with HPP often exhibit a decreased ability or inability to perform routine activities that healthy patients perform on a daily basis without requiring assistance.

Notably, the treatment of HPP, particularly the physical impairments associated with HPP, for an extended period of time, is unknown. Furthermore, the efficacy of HPP treatments to alleviate pain associated with HPP is unknown. Thus, there exists a need for methods that can be used to treat HPP in patients, such as children having HPP, for extended durations so that these patients can live with decreased physical impairments and decreased pain.

SUMMARY

The disclosure relates to methods to identify children having hypophosphatasia (HPP; e.g., children of about 5 to about 12 years of age having HPP) for treatment with a soluble alkaline phosphatase (sALP; such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa). Exemplary tests useful in the methods include (1) the Bruininks-Oseretsky Test of Motor Proficiency 2nd Edition (BOT-2), (2) the Childhood Health Assessment Questionnaire (CHAO), (3) the Pediatric Outcomes Data Collection Instrument (PODCI), (4) the Six Minute Walk Test (6MWT), (5) the Bayley Scales of Infant and Toddler Development, 3rd Edition (BSID-III), and (6) the Peabody Developmental Motor Scales, 2nd Edition (PDMS-2).

The methods further include the use of one or more of the described tests (e.g., the BOT-2, the CHAQ, the 6MWT, the BSID-III, and/or the PDMS-2) singly or in combination to assess treatment efficacy using a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) for a patient having HPP (e.g., a child of about 5 to about 12 years of age having HPP), in which improvements relative to a certain test score demonstrate that the sALP is effective for treating HPP. Additionally, methods may further include changing the dosage and/or frequency of sALP in order to determine the effective amount of the sALP to administer to a child having HPP (e.g., a child of about 5 to about 12 years of age).

A first aspect features a method of treating HPP in a child of about 5 to about 12 years of age having an average Bruininks-Oseretsky Test of Motor Proficiency 2nd Edition (BOT-2) score of less than about 10 (e.g., an average BOT-2 strength score, an average BOT-2 running speed and agility score, and/or an average BOT-2 composite strength and agility score), which includes administering a soluble alkaline phosphatase (sALP) to the child at a dosage providing about 6 mg/kg/week of the sALP (e.g., asfotase alfa). The sALP may have an amino acid sequence with at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 1. Administration of the sALP (e.g., asfotase alfa; SEQ ID NO: 1) results in an average increase in the BOT-2 strength score to about 10 to about 20 after a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)).

The method includes using the BOT-2 score (e.g., an average BOT-2 strength score, an average BOT-2 running speed and agility score, and/or an average BOT-2 composite strength and agility score) to assess the child having HPP prior to administration of the sALP such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa).

In particular, the method can feature treating HPP in a child of about 5 to about 12 years of age having an average BOT-2 strength score of less than 10, which includes administering a soluble alkaline phosphatase (sALP) to the child at a dosage providing about 6 mg/kg/week of the sALP (e.g., asfotase alfa). The sALP includes an amino acid sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 1. Administration of the sALP (e.g., asfotase alfa; SEQ ID NO: 1) results in an average increase in the BOT-2 strength score to about 10 to about 20 after a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)).

The average BOT-2 strength score of the child of the first aspect may be determined relative to a BOT-2 strength score of a child without HPP. In particular, the average BOT-2 strength score of the child may be determined from measurements of activities selected from the group consisting of sit-ups, V-ups, standing long jump, wall sit, and push-ups.

The child having an average BOT-2 strength score of less than 10 exhibits physical impairments relative to the child without HPP. Furthermore, the child exhibits a decrease in physical impairments after administration of the sALP (e.g., asfotase alfa; SEQ ID NO: 1), in which the decrease in physical impairments is sustained throughout administration of the sALP (e.g., asfotase alfa; SEQ ID NO: 1) to the child. Additionally, the average BOT-2 strength score of the child increases to about 12 to about 16 after treatment with the sALP.

Additionally, the method can feature treating HPP in a child of about 5 to about 12 years of age having an average BOT-2 running speed and agility score of less than 5 by administering a sALP (e.g., asfotase alfa; SEQ ID NO: 1) to the child at a dosage providing about 6 mg/kg/week of the sALP. The sALP may have an amino acid sequence with at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 1. Administration of the sALP (e.g., asfotase alfa; SEQ ID NO: 1) results in an average increase in the BOT-2 running speed and agility score to about 5 to about 20 after a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)).

The average BOT-2 running speed and agility score of the child of the second aspect may be determined relative to a BOT-2 running speed and agility score of a child without HPP. In particular, the average BOT-2 running speed and agility score of the child may be determined from measurements of activities selected from the group consisting of stepping over a balance beam, shuttle run, two-legged side hop, and one-legged side hop.

The child having an average BOT-2 running speed and agility score of less than 5 exhibits physical impairments relative to the child without HPP. Furthermore, the child exhibits a decrease in physical impairments after administration of the sALP (e.g., asfotase alfa; SEQ ID NO: 1), in which the decrease in physical impairments is sustained throughout administration of the sALP to the child. Additionally, the average BOT-2 running speed and agility score of the child increases to about 9 to about 13 after treatment with the sALP.

A second aspect features a method of treating HPP in a child of about 5 to about 12 years of age having an average Childhood Health Assessment Questionnaire (CHAQ) disability index and/or discomfort score of greater than about 0.8 by administering a sALP (e.g., asfotase alfa; SEQ ID NO: 1) to the child at a dosage providing about 6 mg/kg/week of the sALP (e.g., asfotase alfa; SEQ ID NO: 1). The sALP may be an amino acid sequence with at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 1. Administration of the sALP (e.g., asfotase alfa; SEQ ID NO: 1) results in an average decrease in the CHAQ disability index and/or discomfort score to about 0 to equal to or less than about 0.5 after a period of at least four (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)). The method includes using the CHAQ disability index and/or discomfort score to assess the child having HPP prior to administration of the sALP such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa).

Additionally, the average CHAQ disability index and/or discomfort score of the child may be determined relative to a CHAQ disability index and/or discomfort score of a child without HPP. The child having an average CHAQ disability index and/or discomfort score of greater than 0.5 exhibits disability in activities of daily living (ADL) and pain relative to the child without HPP. For instance, the child exhibits an increase in ADL after administration of the sALP (e.g., asfotase alfa; SEQ ID NO: 1). The increase in ADL may be sustained throughout administration of the sALP (e.g., asfotase alfa; SEQ ID NO: 1) to the child. For example, administration of the sALP (e.g., asfotase alfa; SEQ ID NO: 1) to the child results in a decrease in pain, as determined using the CHAQ disability index and/or discomfort score. The decrease in pain is sustained throughout administration of the sALP to the child. Furthermore, the average CHAQ disability index and/or discomfort score of the child may decrease to about 0 to equal to or less than about 0.25.

A third aspect features a method of treating HPP in a child of about 5 to about 12 years of age having an average Pediatric Outcomes Data Collection Instrument (PODCI) transfer and mobility, sports and physical functioning, and/or pain score of less than about 40 by administering a sALP to the child at a dosage providing about 6 mg/kg/week of the sALP (e.g., asfotase alfa; SEQ ID NO: 1). The sALP may have an amino acid sequence with at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 1. Administration of the sALP (e.g., asfotase alfa; SEQ ID NO: 1) results in an average increase in the PODCI transfer and mobility, sports and physical functioning, and/or pain score to about 40 to about 50 after a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)). The method includes using the PODCI transfer and mobility, sports and physical functioning, and/or pain score to assess the child having HPP prior to administration of the sALP such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa).

Additionally, the average PODCI transfer and mobility, sports and physical functioning, and/or pain score of the child of the third aspect may be determined relative to a PODCI transfer and mobility, sports and physical functioning, and/or pain score of a child without HPP. The child having an average PODCI transfer and mobility, sports and physical functioning, and/or pain score of less than 40 exhibits disability in ADL and pain relative to the child without HPP. For instance, administration of the sALP (e.g., asfotase alfa; SEQ ID NO: 1) to the child results in an increase in ADL, as determined using the PODCI transfer and mobility, sports and physical functioning, and/or pain score. The increase in ADL is sustained throughout administration of the sALP to the child. For example, the child may also exhibit a decrease in pain after administration of the sALP (e.g., asfotase alfa; SEQ ID NO: 1). The decrease in pain is sustained throughout administration of the sALP to the child.

A fourth aspect features a method of treating HPP in a child of about 5 to about 12 years of age by administering a sALP to the child at a dosage providing about 6 mg/kg/week of the sALP (e.g., asfotase alfa; SEQ ID NO: 1). The sALP can have an amino acid sequence with at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 1. Administration of the sALP for a treatment period of at least one year (e.g., at least two years, at least three years, at least four years, at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)) results in an average increase in a Six Minute Walk Test (6MWT) distance of the child relative to the 6MWT distance of the child prior to administration of the sALP.

For example, the average increase in the 6MWT distance is about 20 meters or greater (e.g., about 20 meters, about 25 meters, about 30 meters, about 35 meters, about 40 meters, about 45 meters, about 50 meters, about 55 meters, about 60 meters, about 65 meters, about 70 meters, about 75 meters, about 80 meters, about 85 meters, about 90 meters, about 95 meters, about 100 meters, or greater). The child can also exhibit an improvement in rickets as determined by the Rickets Severity Scale (RSS) after administration of the sALP. Additionally, the child can exhibit an increase in ADL after administration of the sALP, in which the ADL is determined by a CHAQ disability index score or a PODCI global function, transfer and mobility, or sports and physical functioning scale score.

In any of the above aspects, the child of about 5 to about 12 years of age exhibited an average Bayley Scales of Infant and Toddler Development, 3rd Edition (BSID-III) scaled score of less than about 2 at about 3 years of age or less than 3 years of age. Administration of the sALP (e.g., asfotase alfa; SEQ ID NO: 1) results in an average increase in the BSID-III scaled score to greater than about 5 after a five year period. The average BSID-III scaled score of the child may be determined relative to an average BSID-III scaled score of a child without HPP. In particular, the child having an average BSID-III scaled score of less than 2 exhibits delayed motor development relative to a child without HPP. For instance, the child exhibits an increase in motor development after administration of the sALP (e.g., asfotase alfa; SEQ ID NO: 1), in which the increase in motor development is sustained throughout administration of the sALP to the child, e.g., the average BSID-III scaled score increases to about 5 to about 7.

The average BSID-III scaled score of the child may be determined from measurements of activities selected from the group consisting of prehension, perceptual-motor integration, motor planning and speed, visual tracking, reaching, object grasping, object manipulation, functional hand skills, responses to tactile information, movement of the limbs and torso, static positioning, dynamic movement, balance, and motor planning.

In any of the above aspects, the child of about 5 to about 12 years of age exhibits an average Peabody Developmental Motor Scales, 2nd Edition (PDMS-2) standard score of about 5 at about 3 years of age or less than 3 years of age. Administration of the sALP (e.g., asfotase alfa; SEQ ID NO: 1) results in an average increase in the PDMS-2 standard score to about 7 after a period of at least four years. The average PDMS-2 standard score of the child may be determined relative to a PDMS-2 standard score of a child without HPP. In particular, the child having an average PDMS-2 standard score of about 5 exhibits delayed motor development relative to the child without HPP. For instance, the child exhibits an increase in motor development after administration of the sALP (e.g., asfotase alfa; SEQ ID NO: 1). The increase in motor development is sustained throughout administration of the sALP to the child.

For example, the average PDMS-2 standard score of the child may be determined from measurements of activities selected from the group consisting of crawling, walking, running, hopping, jumping forward, reflexes, balance, object manipulation, grasping, and visual-motor integration.

In any of the above aspects, the sALP (e.g., asfotase alfa; SEQ ID NO: 1) is formulated for daily or weekly administration, e.g., the sALP (e.g., asfotase alfa; SEQ ID NO: 1) is formulated for administration twice a week, three time a week, four times a week, five times a week, six times a week, or seven times a week. For example, the sALP (e.g., asfotase alfa; SEQ ID NO: 1) is formulated at a dosage of 2 mg/kg for administration three times a week, the sALP (e.g., asfotase alfa; SEQ ID NO: 1) is formulated at a dosage of 1 mg/kg for administration six times a week, or the sALP (e.g., asfotase alfa; SEQ ID NO: 1) is formulated at a dosage of 3 mg/kg for administration three times a week.

For any of the above aspects, the sALP (e.g., asfotase alfa; SEQ ID NO: 1) is administered for at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or for more than ten years (e.g., the sALP is administered for the lifetime of the patient).

In any of the above aspects, the sALP (e.g., asfotase alfa) includes or consists of the amino acid sequence of SEQ ID NO: 1.

For any of the above aspects, the method may further include determining sALP activity (e.g., asfotase alfa activity). The determination of sALP activity may include measuring at least one of phosphoethanolamine (PEA), inorganic pyrophosphate (PPi), and/or pyridoxal 5′-phosphate (PLP) in a serum and/or blood sample from the child.

In any of the above aspects, the sALP (e.g., asfotase alfa; SEQ ID NO: 1) is administered in an amount that is therapeutically effective to treat at least one symptom of HPP (e.g., rickets, premature loss of deciduous teeth, incomplete bone mineralization, elevated blood and/or urine levels of inorganic pyrophosphate (PR), elevated blood and/or urine levels of phosphoethanolamine (PEA), elevated blood and/or urine levels of pyridoxal 5′-phosphate (PLP), hypomineralization, rachitic ribs, hypercalciuria, short stature, skeletal deformity, waddling gait, bone pain, bone fracture, HPP-related seizure, inadequate weight gain, and/or calcium pyrophosphate dihydrate crystal deposition).

For any of the above aspects, the sALP (e.g., asfotase alfa; SEQ ID NO: 1) may be administered at an initial dosage of about 3 mg/kg/week and then increased to a dosage of about 6 mg/kg/week or more. Furthermore, the dosage may be increased to about 6 mg/kg/week or more after about three months to about nine months. Additionally, the dosage may be increased to about 9 mg/kg/week.

For any of the above aspects, the child may exhibit symptoms of HPP at birth or after birth (e.g., at five years of age or older).

In any of the above aspects, the child exhibits tolerability to administration of the sALP (e.g., asfotase alfa; SEQ ID NO: 1). For instance, the tolerability includes a lack or decreased incidence of adverse events selected from the group consisting of injection site erythema, decrease in hemoglobin, pyrexia, pneumonia, upper respiratory tract infection, craniosynostosis, otitis media, vomiting, constipation, diarrhea, tooth loss, nasopharyngitis, rash, dental carries, and irritability.

In any of the above aspects, the child may be one that does not exhibit serum calcium and/or phosphorus levels below the age-adjusted normal range. Additionally, the child may be one that does not exhibit symptoms of a treatable form of rickets. Furthermore, the child may be one that has not previously received treatment with a bisphosphonate.

For any of the above aspects, the method further includes performing radiographs of the child to determine an average Radiographic Global Impression of Change (RGI-C) score, in which the child exhibits an average RG1-C score of less than 2 prior to administration of the sALP (e.g., asfotase alfa; SEQ ID NO: 1). In particular, the average RGI-C score of the child may be determined relative to an average RGI-C score of a child without HPP. Preferably, the child exhibits increased bone density after administration of the sALP (e.g., asfotase alfa; SEQ ID NO: 1). The increase in bone density is sustained throughout administration of the sALP. For instance, the child exhibits an average RGI-C score of greater than 2.

For any of the above aspects, the method further includes determining weight and/or length of the child, in which a Z-score is determined from the weight and/or length of the child. Preferably, the child exhibits an improved Z-score for weight and/or length after administration of the sALP (e.g., asfotase alfa; SEQ ID NO: 1).

In any of the above aspects, the sALP (e.g., asfotase alfa; SEQ ID NO: 1) is formulated in a pharmaceutical composition, e.g., with a pharmaceutically acceptable carrier. For example, the pharmaceutically acceptable carrier is saline. The pharmaceutically acceptable carrier may include sodium chloride and sodium phosphate (e.g., the pharmaceutically acceptable carrier may contain about 150 mM sodium chloride and about 25 mM sodium phosphate).

In any of the above aspects, the pharmaceutical composition is formulated for subcutaneous, intramuscular, intravenous, oral, nasal, sublingual, intrathecal, or intradermal administration. In particular, the pharmaceutical composition is formulation for subcutaneous administration.

For any of the above aspects, the sALP (e.g., asfotase alfa; SEQ ID NO: 1) is physiologically active toward PEA, PPi, and PLP. For example, the sALP (e.g., asfotase alfa; SEQ ID NO: 1) is catalytically competent to improve skeletal mineralization in bone. Preferably, the sALP (e.g., asfotase alfa; SEQ ID NO: 1) is the soluble extracellular domain of an alkaline phosphatase.

In any of the above aspects, the child has not previously been administered the sALP (e.g., asfotase alfa; SEQ ID NO: 1).

Additionally, the child may have been continuously treated with the sALP (e.g., asfotase alfa; SEQ ID NO: 1), according to the treatment regimens described herein for a period of days (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, or more days), weeks (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 45, 50, or more weeks), months (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months), or years (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, or more years). Alternatively, the child may have been continuously treated with the sALP (e.g., asfotase alfa; SEQ ID NO: 1) for a period of days, weeks, months, or years, and then treatment with the sALP was discontinued for a period of days (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, or more days), weeks (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 45, 50, or more weeks), months (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months), or years (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, or more years).

Definitions

As used herein, “a” or “an” means “at least one” or “one or more” unless otherwise indicated. In addition, the singular forms “a”, “an”, and “the” include plural referents unless the context clearly dictates otherwise.

As used herein, “about” refers to an amount that is ±10% of the recited value and is preferably ±5% of the recited value, or more preferably ±2% of the recited value.

As used herein, “at least” refers to an amount that is ≤10% of the recited value and is preferably ≤5% of the recited value, or more preferably ≤2% of the recited value.

By “asfotase alfa” is meant a human TNALP (hTNALP) fusion protein formulated for the treatment of HPP. Asfotase alfa (STRENSIQ®, Alexion Pharmaceuticals, Inc.) is a fusion protein including a soluble glycoprotein of two identical polypeptide chains, in which each polypeptide chain includes amino acid residues 1-726 of SEQ ID NO: 1. The structure of each polypeptide chain includes the catalytic domain of hTNALP, the human immunoglobulin G1 Fc domain, and a deca-aspartate peptide used as a bone targeting domain (the structure hTNALP-Fc-D10). The two polypeptide chains are covalently linked by two disulfide bonds. Asfotase alfa has been approved under the trade name STRENSIQ® in the United States, Europe, Japan, Canada, Israel, Australia, and Korea.

As used herein, “average” refers to a numerical value expressing the mean or median of a data set. The mean of a data set is calculated by dividing the sum of the values in the set by their number. The median of a date set is calculated by determining the middle value in a list of odd numbers or by determining the mean of the two data values in the middle in a list of even numbers.

The terms “Bayley Scales of Infant and Toddler Development, 3rd Edition” or “BSID-III” as used herein refer to a standardized series of measurements used to assess the motor (fine and gross), language (receptive and expressive), and cognitive development of patients, e.g., infants and toddlers. See Bayley, (2006). Bayley scales of infant and toddler development: administration manual. San Antonio, Tex.: Harcourt Assessment, hereby incorporated by reference in its entirety. In particular, the BSID-III is designed to assess infants and young children. The BSID-III measurements include a series of developmental play tasks to be administered to the patient. Raw scores of successfully completed items are converted to scaled scores. The scaled scores are then used to determine the patient's performance compared to healthy, age-adjusted patients. The BSID-III can also include the Social-Emotional Adaptive Behavior Questionnaire, which is completed by the parent/guardian, to establish the range of adaptive behaviors of the patient. For example, measurements for determining the BSID-III score (e.g., the BSID-III gross motor function score) can include prehension, perceptual-motor integration, motor planning and speed, visual tracking, reaching, object grasping, object manipulation, functional hand skills, responses to tactile information, movement of the limbs and torso, static positioning, dynamic movement, balance, and motor planning. These patient measurements are then converted into a BSID-III scaled score (e.g., the BSID-III gross motor function scaled score) ranging from 0 to 14, in which scores of about 7 to about 13 are considered the normal range of healthy patients.

The term “bone-targeting moiety,” as used herein, refers to an amino acid sequence of between 1 and 50 amino acid residues in length having a sufficient affinity to the bone matrix, such that the bone-targeting moiety, singularly, has an in vivo binding affinity to the bone matrix that is about 10−6 M to about 10−15 M (e.g., 10−7 M, 10−8 M, 10−9 M, 10−10 M, 10−11 M, 10−12 M, 10−13 M, 10−14 M, or 10−15 M).

The terms “Bruininks-Oseretsky Test of Motor Proficiency 2nd Edition” or “BOT-2,” as used herein, refer to the second edition of a standardized test of gross and fine motor performance for patients, e.g., from about 4 to about 21 years of age. See Bruininks, R. H. (2005). Bruininks-Oscretsky Test of Motor Proficiency, (BOT-2). Minneapolis, Minn.: Pearson Assessment, hereby incorporated by reference in its entirety. The BOT-2 is administered individually to assess gross and fine motor skills of a range of patients. In particular, the BOT-2 can be used to evaluate physical impairments and mobility restrictions in patients having HPP. The BOT-2 provides composite BOT-2 scores in the following areas: strength, running speed and agility, fine motor precision, fine motor integration, manual dexterity, bilateral coordination, balance, and upper-limb coordination. For example, a BOT-2 strength score can be determined by having a patient perform sit-ups, v-ups, standing long jump, wall sit, and push-ups. A running speed and agility score can be determined by having a patient step over a balance beam or perform a shuttle run, two-legged side hop, or one-legged side hop. Both BOT-2 strength and BOT-2 running speed and agility scores range from 0 to 25, in which a score of about 10 to 20 is considered representative of healthy patients.

The terms “Childhood Health Assessment Questionnaire” or “CHAQ,” as used herein refer to a questionnaire that is used to assess the health status (e.g., ability to perform activities of daily living (ADLs) and incidence of pain) of patients of 1 to 19 years of age, such as patients with HPP. For a description of the CHAQ test and index, see Bruce & Fries (J. Rheumatol. 30(1): 167-178, 2003), hereby incorporated by reference in its entirety. The CHAQ may be administered by interview or self-report for children greater than 8 years of age. The CHAQ includes eight sub-scales for dressing/grooming, arising, eating, walking, hygiene, reach, grip, and activities. The range of scores within each category is from 0 to 3, in which a score of 0 indicates without any difficulty; a score of 1 indicates with some difficulty; a score of 2 indicates with much difficulty; and a score of 3 indicates that the patient is unable to perform the activity. The CHAQ index may also be used to determine the presence and severity of pain.

By “extracellular domain” is meant any functional extracellular portion of the native protein, e.g., alkaline phosphatase. In particular, the extracellular domain lacks the signal peptide.

By “Fc” is meant a fragment crystallizable region of an immunoglobulin, e.g., IgG-1, IgG-2, IgG-3, IgG-3 or IgG-4, including the CH2 and CH3 domains of the immunoglobulin heavy chain. Fc may also include any portion of the hinge region joining the Fab and Fc regions. The Fc can be of any mammal, including human, and may be post-translationally modified (e.g., by glycosylation). In a non-limiting example, Fc can be the fragment crystallizable region of human IgG-1 having the amino acid sequence of SEQ ID NO: 20.

By “fragment” is meant a portion of a polypeptide or nucleic acid molecule that contains, preferably, at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or more of the entire length of the reference nucleic acid molecule or polypeptide. A fragment may contain, e.g., 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 400, 500, 600, 700, or more amino acid residues, up to the entire length of the polypeptide. Exemplary sALP fragments have amino acid residues 18-498, 18-499, 18-500, 18-501, 18-502, 18-503, 18-504, 18-505, 18-506, 18-507, 18-508, 18-509, 18-510, 18-511, or 18-512 of a ALP (e.g., SEQ ID NOs: 2-6), and may include additional C-terminal and/or N-terminal portions.

The terms “hypophosphatasia” or “HPP,” as used herein, refer to a rare, heritable skeletal disorder caused by, e.g., one or more loss-of-function mutations in the ALPL (alkaline phosphatase, liver/bone/kidney) gene, which encodes tissue-nonspecific alkaline phosphatase (TNALP). HPP may be further characterized as infantile HPP, childhood HPP, perinatal HPP (e.g., benign perinatal HPP or lethal perinatal HPP), or odonto-HPP.

The term “HPP phenotype,” as used herein, refers to any one of rickets (defect in growth plate cartilage), osteomalacia, elevated blood and/or urine levels of inorganic pyrophosphate (PP;), phosphoethanolamine (PEA), or pyridoxal 5′-phosphate (PLP), seizure, bone pains, and calcium pyrophosphate dihydrate crystal deposition (CPPD) in joints leading to chondrocalcinosis, craniosynostosis, and premature death. Without being so limited, a HPP phenotype can be documented by one or more of growth retardation with a decrease of long bone length (including but not limited to femur, tibia, humerus, radius, and/or ulna), a decrease of the mean density of total bone and a decrease of bone mineralization in bones such as femur, tibia, ribs and metatarsi, and phalange, a decrease in teeth mineralization, and a premature loss of deciduous teeth (e.g., aplasia, hypoplasia, or dysplasia of dental cementum). Without being so limited, correction or prevention of bone mineralization defect may be observed by one or more of the following: an increase of long bone length, an increase of mineralization in bone and/or teeth, a correction of bowing of the legs, a reduction of bone pain and a reduction of CPPD crystal deposition in joints.

By “naïve patient” is meant a patient having HPP (e.g., a child) that has never received treatment with a sALP such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa).

By “pain” as used herein refers to physical suffering or discomfort caused by HPP, such as bone pain. For instance, symptoms of pain can include, e.g., soreness, tightness, or stiffness. The severity of pain can vary between patients (e.g., chronic pain or acute pain). In particular, chronic pain refers to pain that lasts longer than three to six months or pain that extend beyond the expected period of healing. In contrast, acute pain refers to pain that typically lasts less than three to six months. As described herein, therapeutic compositions (e.g., including a sALP, such as asfotase alfa) can be administered to a patient suffering from pain (e.g., bone pain) in an amount sufficient to relieve or at least partially relieve the symptoms of pain (e.g., discomfort, soreness, tightness, or stiffness) and its complications (e.g., fatigue, sleeplessness, weakened immune system, depression, anxiety, stress, irritability, or disability).

The terms “peptide,” “polypeptide,” and “protein” are used interchangeably and refer to any chain of two or more natural or unnatural amino acid residues, regardless of post-translational modification (e.g., glycosylation or phosphorylation), constituting all or part of a naturally-occurring or non-naturally occurring polypeptide or peptide, as is described herein.

By “pharmaceutically acceptable carrier” or “pharmaceutically acceptable excipient” is meant at least one carrier or excipient, respectively, which is physiologically acceptable to the treated patient while retaining the therapeutic properties of the compound with which it is administered. One exemplary pharmaceutically acceptable carrier substance is physiological saline. For instance, the pharmaceutically acceptable carrier can include sodium chloride (e.g., 150 mM sodium chloride) and sodium phosphate (e.g., 25 mM sodium phosphate). Other physiologically acceptable carriers and their formulations are known to those skilled in the art and described, e.g., in Remington's Pharmaceutical Sciences (20th edition), A. Gennaro, Ed., 2000, Lippincott, Williams & Wilkins, Philadelphia, Pa.

By “pharmaceutical composition” is meant a composition containing a polypeptide or nucleic acid molecule as described herein formulated with at least one pharmaceutically acceptable excipient, diluent, or carrier. The pharmaceutical composition may be manufactured or sold with the approval of a governmental regulatory agency as part of a therapeutic regimen for the treatment or prevention of a disease or event in a patient. Pharmaceutical compositions can be formulated, for example, for subcutaneous administration, intravenous administration (e.g., as a sterile solution free of particulate emboli and in a solvent system suitable for intravenous use), for oral administration (e.g., a tablet, capsule, caplet, gelcap, or syrup), or any other formulation described herein, e.g., in unit dosage form.

The term “physical impairments,” as used herein, refers to a physiological condition, such as bone weakness and muscle weakness, associated with HPP that can restrict or eliminate, e.g., ambulation, functional endurance, and ability to perform activities of daily living (ADL) of a patient. In particular, physical impairments may restrict or eliminate a patient's ability to perform ADL, which are routine activities that healthy patients perform on a daily basis without requiring assistance, such as functional mobility or transferring (e.g., walking), bathing and showering, dressing, self-feeding, and personal hygiene and grooming. As described herein, therapeutic compositions (e.g., compositions including a sALP, such as asfotase alfa) can be administered to a patient to decrease the severity and/or frequency of physical impairments associated with an HPP phenotype.

The terms “Pediatric Outcomes Data Collection Instrument” or “PODCI,” as used herein, refer to a questionnaire used to assess overall health, incidence of pain, and ability to perform ADLs of patients under 19 years of age, particularly in patients with chronic health disorders, such as patients with HPP. For a description of the PODCI, see Plint et al. (J. Pediatr. Orthop. 23(6): 788-790, 2003), hereby incorporated by reference in its entirety. The questionnaire may be completed by the patient or by a parent/guardian of the patient with knowledge of the patient's condition. The eight scales generated from the PODCI include the following: 1) the upper extremity and physical function scale to measure difficulty encountered in performing daily personal care and student activities; 2) the transfer and basic mobility scale to measure difficulty experienced in performing routine motion and motor activities in daily activities; 3) the sports/physical functioning scale to measure difficulty or limitations encountered in participating in more active activities or sports; 4) the pain/comfort scale to measure the level of pain experienced during the past week; 5) the treatment expectations scale to measure the long term expectations of treatment; 6) the happiness scale to measure overall satisfaction with personal looks and sense of similarity to friends and others of own age; 7) the satisfaction with symptoms scale to measure the patients acceptance of current limitations should this be a life-long state; and 8) the global functioning scale, which is a general combined scale calculated from the first four scales listed above. Standardized scores are generated from a series of questions in the PODCI and converted to a 0 to 100 scale, in which 0 represents significant disability and 100 represents less disability.

The terms “Peabody Developmental Motor Scales, 2nd Edition” or “PDMS-2,” as used herein, refer to an early childhood motor development program that provides an assessment of gross and fine motor skills in patients from birth throughout childhood (e.g., infants and children). For a description of the PDMS-2 scales, see van Hartingsveldt, et al. (Occup. Ther. Int 12(1): 1-13, 2005), hereby incorporated by reference in its entirety. The PDMS-2 is composed of six subtests that measure interrelated motor abilities of early development. The six subtests include the following: 1) the locomotor subtest to measures a patient's ability to move from one place to another (measurements include crawling, walking, running, hopping, and jumping forward); 2) the reflexes subtest to measure a patients ability to automatically react to environmental events; 3) the stationary subtest to measure a patient's ability to sustain control of his or her body within the center of gravity and retain equilibrium; 4) the object manipulation subtest to measure a patient's ability to manipulate an object, such as catching, throwing, and kicking a ball; 5) the grasping subtest to measure a patient's ability to use his or her hands, such as the ability to hold an object with one hand and actions involving the controlled use of the fingers of both hands; and 6) the visual-motor integration subtest to measure a patient's ability to use his or her visual perceptual skills to perform complex eye-hand coordination tasks, such as reaching and grasping for an object, building with blocks, and copying designs. The PDMS-2 measurements for each subtest is converted into a PDMS-2 score, such as the PDMS-2 locomotor standard score ranging from 0 to 13, in which the range of health patients is from about 7 to about 13.

The terms “sALP,” “soluble alkaline phosphatase,” and “extracellular domain of an alkaline phosphatase” are used interchangeably and refer to a soluble, non-membrane-bound alkaline phosphatase or a domain, biologically active fragment, or biologically active variant thereof. sALPs include, for example, an alkaline phosphatase lacking a C-terminal glycolipid anchor (GPI signal sequence, e.g., polypeptides including or consisting of the amino acid residues 18-502 of a human TNALP (SEQ ID NOs: 2, 3, 4, 5, or 6)). In particular, a TNALP may include, e.g., a polypeptide including or consisting of amino acid residues 1-485 of SEQ ID NO: 1, such as asfotase alfa, or a polypeptide variant having at least 95% sequence identity to the amino acid residues 1-485 of SEQ ID NO: 1. sALPs further include, for example, mammalian orthologs of human TNALP, such as a rhesus TNALP (SEQ ID NO: 7), a rat TNALP (SEQ ID NO: 8), a canine TNALP (SEQ ID NO: 9), a porcine TNALP (SEQ ID NO: 10), a murine TNALP (SEQ ID NO: 11), a bovine TNALP (SEQ ID NOs: 12-14), or a feline TNALP (SEQ ID NO: 15). sALPs also include soluble, non-membrane-bound forms of human PALP (e.g., polypeptides including or consisting of amino acid residues 18-502 of SEQ ID NOs: 16 or 17), GCALP (e.g., polypeptides including or consisting of amino acid residues 18-502 of SEQ ID NO: 18), and IALP (e.g., polypeptides including or consisting of amino acid residues 18-502 of SEQ ID NO: 19), and additional variants and analogs thereof that retain alkaline phosphatase activity, e.g., the ability to hydrolyze PPi. A sALP, in particular, lacks the N-terminal signal peptide (e.g., aa 1-17 of SEQ ID NOs: 2-6, 8, 11-13, or 15 or aa 1-25 of SEQ ID NO: 7).

By “sALP polypeptide” is meant a polypeptide having the structure A-sALP-B, wherein sALP is as defined herein and each of A and B is absent or is an amino acid sequence of at least one amino acid (e.g., any sALP fusion polypeptide described herein (for example the sALP fusion polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa).

By “signal peptide” is meant a short peptide (5-30 amino acids long) at the N-terminus of a polypeptide that directs a polypeptide towards the secretory pathway (e.g., the extracellular space). The signal peptide is typically cleaved during secretion of the polypeptide. The signal sequence may direct the polypeptide to an intracellular compartment or organelle, e.g., the Golgi apparatus. A signal sequence may be identified by homology, or biological activity, to a peptide with the known function of targeting a polypeptide to a particular region of the cell. One of ordinary skill in the art can identify a signal peptide by using readily available software (e.g., Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, Wis. 53705, BLAST, or PILEUP/PRETTYBOX programs). A signal peptide can be one that is, for example, substantially identical to amino acid residues 1-17 of SEQ ID NOs: 2-6 or amino acid residues 1-25 of SEQ ID NO: 7.

As used herein, when a polypeptide or nucleic acid sequence is referred to as having “at least X % sequence identity” to a reference sequence, wherein “X” is a real number, it is meant that at least X percent of the amino acid residues or nucleotides in the polypeptide or nucleic acid are identical to those of the reference sequence when the sequences are optimally aligned. An optimal alignment of sequences can be determined in various ways that are within the skill in the art, for instance, the Smith Waterman alignment algorithm (Smith et al., J. Mol. Biol. 147:195-7, 1981) and BLAST (Basic Local Alignment Search Tool; Altschul et al., J. Mol. Biol. 215: 403-10, 1990). These and other alignment algorithms are accessible using publicly available computer software such as “Best Fit” (Smith and Waterman, Advances in Applied Mathematics, 482-489, 1981) as incorporated into GeneMatcher Plus (Schwarz and Dayhoff, Atlas of Protein Sequence and Structure, Dayhoff, M. O., Ed pp 353-358, 1979), BLAST, BLAST-2, BLAST-P, BLAST-N, BLAST-X, WU-BLAST-2, ALIGN, ALIGN-2, CLUSTAL, Megalign (DNASTAR), or other software/hardware for alignment. In addition, those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve optimal alignment over the length of the sequences being compared.

The term “patient” refers to a mammal, including, but not limited to, a human or a non-human mammal, such as a bovine, equine, canine, ovine, or feline.

As used herein, “Six Minute Walk Test” and “6MWT” refer to a standardized test to assess walking ability of a patient (e.g., a child having HPP), in particular, the ability of the patient to lift and set down each foot in turn. See the American Thoracic Society statement: guidelines for the six-minute walk test (American Journal of Respiratory and Critical Care Medicine, 166(1):111-7, 2002), hereby incorporated by reference in its entirety. The 6MWT is determined from the distance (e.g., in meters) that a patient walks on a flat, hard surface in a period of six minutes. The 6MWT distance can then be compared to the 6MWT distance of the patient at baseline, the 6MWT distance of an untreated subject (e.g., an untreated subject of about the same age, height, and/or gender), or the 6MWT distance of a healthy subject (e.g., a healthy subject of about the same age, height, and/or gender) and expressed as a percentage to determine the 6MWT value.

By “therapeutically effective amount” is meant an amount of a polypeptide or nucleic acid molecule described herein that is sufficient to substantially improve, treat, prevent, delay, suppress, or arrest at least one symptom of HPP. A therapeutically effective amount of a composition described herein may depend on the severity of the disorder being treated and the condition, weight, and general state of the patient and can be determined by an ordinarily-skilled artisan with consideration of such factors. A therapeutically effective amount of a composition described herein can be administered to a patient in a single dose or in multiple doses administered over a period of time.

By “treating,” “treat,” or “treatment” is meant the medical management of a patient with the intent to cure, ameliorate, stabilize, reduce the likelihood of, or prevent HPP and/or management of a patient exhibiting or likely to have HPP, e.g., by administering a pharmaceutical composition. This term includes active treatment, that is, treatment directed specifically toward the improvement or associated with the cure of a disease, pathological condition, disorder, or event, and also includes causal treatment, that is, treatment directed toward removal of the cause of the associated disease, pathological condition, disorder, or event. In addition, this term includes palliative treatment, that is, treatment designed for the relief or improvement of at least one symptom rather than the curing of the disease, pathological condition, disorder, or event; symptomatic treatment, that is, treatment directed toward constitutional symptoms of the associated disease, pathological condition, disorder, or event; preventative treatment, that is, treatment directed to minimizing or partially or completely inhibiting the development of the associated disease, pathological condition, disorder, or event, e.g., in a patient who is not yet ill, but who is susceptible to, or otherwise at risk of, a particular disease, pathological condition, disorder, or event; and supportive treatment, that is, treatment employed to supplement another specific therapy directed toward the improvement of the associated disease, pathological condition, disorder, or event.

Other features and advantages of the present disclosure will be apparent from the following Detailed Description, the drawings, and the claims.

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1 is an image showing the study design for administering asfotase alfa to children with hypophosphatasia (HPP) over a time period of 5 years, including an initial phase of treatment with asfotase alfa, from baseline to 6 months, and an extension phase of treatment with asfotase alfa (from 6 months of treatment to 5 or more years of treatment). During the 6 month initial phase involving 13 participants, 1 child withdrew for elective surgery after 1 month of treatment. The remaining 12 were assessed up to 5 years (60 months) of treatment and continue on study. Data were pooled across these 2 phases for analysis. The radiographic findings were contrasted to 2 year experience with 16 historical control patients. LOCF, last observation carried forward.

FIG. 2 is a radiographic image showing skeletal features of HPP in a child prior to treatment with asfotase alfa. Untreated, the radiographic features of hypophosphatasia in children include, as in the wrist pictured, osteopenia, metaphyseal fraying, metaphyseal flaring, and metadiaphyseal sclerosis, and as in the knee pictured, characteristic tongues of radiolucency, irregularity of the provisional zone of calcification, distal metaphyseal demineralization, transverse subphyseal band of lucency, and apparent physeal widening.

FIG. 3 is a graph showing changes in the Radiographic Global Impression of Change (RGI-C) scores of HPP children after administration of asfotase alfa over a treatment period of 6 months and 2 years relative to historical untreated controls. Individual dots illustrate the distribution of individual patient scores at each time point. Boxes represent the median RGI-C scores and first and third quartiles. Whisker lines above and below the boxes represent the end-range of patient scores. The RGI-C is scored on a scale ranging from −3 to +3, with 0 representing no change. Negative values represent worsening, and positive values represent improvement or healing. A score of +3 indicates nearly complete or complete healing. Median RGI-C scores for the historical controls and asfotase alfa were 0 and 2, respectively, at 6 months and 2 years. The number of patients assessed at each time point is shown below each bar. P≤0.0001 by Wilcoxon signed-rank test at all time points compared with no change.

FIG. 4 is a series of images showing representative radiographic changes of a child with HPP at baseline and after 6 months, 3 years, and 5 years of treatment with asfotase alfa. The child was 6 years old at baseline. RGI-C and Rickets Severity Score (RSS) scores over time are also shown.

FIGS. 5A-5C are graphs showing alkaline phosphatase (ALP) activity (FIG. 5A), pyridoxal 5′-phosphate (PLP) concentrations (FIG. 5B), and inorganic pyrophosphate (PPi) concentrations (FIG. 5C) in samples from HPP children after administration of asfotase alfa over a time period of 5 years. PLP and PPi concentrations are shown relative to the normal range for healthy children. Median, min, and max values are given below each panel.

FIGS. 6A-6C are graphs showing growth during treatment of HPP children with asfotase alfa over a time period of 5 years. Growth was assessed for weight Z-scores (FIG. 6A), height Z-scores (FIG. 6B), and body mass index (BMI) Z-scores (FIG. 6C). For all graphs, individual dots indicate individual patient scores at each time point. Median, min, max, and n values are shown below each panel. P-values at 5 years are from a Wilcoxon signed-rank test.

FIGS. 7A-7D are graphs showing right hip abduction (FIG. 7A), right hip extension (FIG. 7B), right knee flexion (FIG. 7C), and right knee extension (FIG. 7D) of HPP children after administration of asfotase alfa over a time period of 5 years. Values are expressed as a percentage of the predicted value for healthy children. For all graphs, individual dots indicate individual patient scores at each time point. Median, min, max, and n values are shown below each panel.

FIGS. 8A-8B are graphs showing individual and median 6MWT values (FIG. 8A) and the percent predicted 6MWT distance median (FIG. 8B) of HPP children administered asfotase alfa over a time period of 5 years. Median, min, max, and n values are shown. Individual dots indicate individual patient scores at each time point. Gray area represents the normal range for the 6MWT distance. P≤0.0005 by paired t test for the mean difference between each time point and baseline.

FIGS. 9A-9B are an image and graph, respectively, of the Bruininks-Oseretsky Test of Motor Proficiency, 2nd Edition (BOT-2) strength tests (FIG. 9A) and the average BOT-2 strength scaled scores of HPP patients administered asfotase alfa over a time period of 5 years (FIG. 9B). The mean±the standard deviation and median of the BOT-2 scores for each time interval (baseline, 6 months, 1 year, 2 years, 3 years, 4 years, and 5 years) are shown. Arrows indicate the initial phase and extension phase, respectively, of treatment with asfotase alfa.

FIGS. 10A-100 are an image and graphs, respectively, of the BOT-2 running speed and agility tests (FIG. 10A) and BOT-2 running speed and agility scaled scores (FIG. 10B) and BOT-2 composite strength and agility standardized scores (FIG. 10C-10D) of HPP patients administered asfotase alfa over a time period of 5 years. The mean±the standard deviation and median of the BOT-2 scores for each time interval (baseline, 6 months, 1 year, 2 years, 3 years, 4 years, and 5 years) are shown. Arrows indicate the initial phase and extension phase, respectively, of treatment with asfotase alfa. Median, min, max, and n values are shown for FIG. 10D.

FIG. 11 is a graph of the Childhood Health Assessment Questionnaire (CHAQ) disability index scores of HPP patients administered asfotase alfa over a time period of 5 years. Average CHAQ disability index scores at baseline, 3 months, 6 months, 1 year, 2 years, 3 years, 4 years, and 5 years are shown. Median CHAQ disability index scores at each time interval are shown in bold. *P≤0.05, **P≤0.01, and ***P≤0.001 mean difference for each time point compared with baseline by paired t test. Median, min, max, and n values are shown. Individual dots indicate individual patient scores at each time point.

FIGS. 12A-12B are graphs of the Pediatric Outcomes Data Collection Instrument (PODCI) median normative scores for transfer and basic mobility (FIG. 12A) and sports/physical functioning (FIG. 12B) of HPP patients administered asfotase alfa over a time period of 5 years. PODCI disability index scores at baseline, 6 months, 1 year, 2 years, 3 years, 4 years, and 5 years are shown. Maximum and median PODCI scores at each time interval are shown in bold. Arrows indicate the initial phase and extension phase, respectively, of treatment with asfotase alfa.

FIGS. 13A-13B are graphs of the median CHAQ discomfort score (FIG. 13A) and median PODCI score (FIG. 13B) of HPP patients after administration of asfotase alfa over a time period of 5 years. Average CHAQ and PODCI scores at baseline, 6 months, 1 year, 2 years, 3 years, 4 years, and 5 years are shown. Maximum and median CHAQ and PODCI scores at each time interval are shown in bold. Arrows indicate the initial phase and extension phase, respectively, of treatment with asfotase alfa.

FIG. 14 is an image showing the study design for the treatment of infants with HPP over a period of 5 years with a asfotase alfa including an initial phase of treatment with asfotase alfa (from baseline to 6 months and an extension phase of treatment with asfotase alfa (from 6 months of treatment to 5 or more years of treatment).

FIGS. 15A-15B are an image and a graph, respectively, showing the RGI-C assessment scale (FIG. 15A) and the average RGI-C scores (FIG. 15B) of HPP patients after administration of asfotase alfa over a time period of 5 years.

FIGS. 16A-16B are graphs showing length Z-scores of HPP patients after administration of asfotase alfa at 0 to 108 months. Male and female patients are shown (FIG. 16A and FIG. 16B, respectively).

FIGS. 17A-17B are graphs showing weight Z-scores of HPP patients after administration of asfotase alfa at 0 to 108 months. Male and female patients are shown (FIG. 17A and FIG. 17B, respectively).

FIG. 18 is a graph showing the median BMI and Z-scores (length and width) of both female and male HPP patients after administration of asfotase alfa at 0 to 108 months.

FIG. 19 is a graph showing the average Bayley Scales of Infant and Toddler Development, 3rd Edition (BSID-III) scores of HPP patients after administration of asfotase alfa over a time period of 3 years. BSID-III scores at baseline, 3 months, 6 months, 1 year, 2 years, and 3 years are shown.

FIG. 20 is a graph showing the average Peabody Developmental Motor Scales, 2nd Edition (PDMS-2) scores of HPP patients after administration of asfotase alfa over a time period of 5 years. PDMS-2 scores at 2 years, 3 years, 4 years, and 5 years are shown.

DETAILED DESCRIPTION

We have discovered that asfotase alfa (SEQ ID NO: 1) can be used effectively to treat hypophosphatasia (HPP), its symptoms, and physical impairments associated therewith in a child with HPP (e.g., a child of about 5 to about 12 years of age having HPP) for an extended period of time (e.g., at least four years, at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)). In particular, asfotase alfa (SEQ ID NO: 1) can be administered to treat children with HPP exhibiting physical impairments (e.g., bone or muscle weakness) and/or pain (e.g., bone or muscle pain). Furthermore, the child having HPP (e.g., a child of about 5 to about 12 years of age having HPP) can be a naïve patient that has not have previously received treatment with asfotase alfa (SEQ ID NO: 1).

Methods for administering asfotase alfa (SEQ ID NO: 1) to a child with HPP (e.g., a child of about 5 to about 12 years of age with HPP) having an average Bruininks-Oseretsky Test of Motor Proficiency 2nd Edition (BOT-2) score indicative of physical impairments (e.g, an average BOT-2 score of less than about 10 in one or more BOT-2 score areas of strength, running speed and agility, fine motor precision, fine motor integration, manual dexterity, bilateral coordination, balance, and upper-limb coordination) are described. For example, asfotase alfa (SEQ ID NO: 1) can be administered to a child with HPP (e.g., a child of about 5 to about 12 years of age having HPP) having an average BOT-2 strength score of less than about 10 and/or an average BOT-2 running speed and agility score of less than about 5. Similarly, methods for administering asfotase alfa (SEQ ID NO: 1) to a child (e.g., a child of about 5 to about 12 years of age having HPP) having a Six Minute Walk Test (6MWT) distance that is indicative of decreased walking ability, e.g., relative to a healthy patient, are also described.

Methods for administering asfotase alfa (SEQ ID NO: 1) to a child (e.g., a child of about 5 to about 12 years of age having HPP) having an average Childhood Health Assessment Questionnaire (CHAQ) index score indicative of disability in activities of daily living (ADL) and pain (e.g., an an average CHAQ index score of greater than about 0.8) are also described. Likewise, methods for administering asfotase alfa (SEQ ID NO: 1) to a child (e.g., a child of about 5 to about 12 years of age having HPP) having an average Pediatric Outcomes Data Collection Instrument (PODCI) score indicative of disability in activities of daily living (ADL) and pain (e.g., an average PODCI score of less than about 40) are also described.

Additionally, methods for administering asfotase alfa (SEQ ID NO: 1) to a child (e.g., a child of about 5 to about 12 years of age having HPP) that exhibited an average Bayley Scales of Infant and Toddler Development, 3rd Edition (BSID-III) score indicative of delayed motor development (e.g., an average BSID-III scaled score of less than 2) at about 3 years of age or less than 3 years of age are described. Likewise, methods for administering asfotase alfa (SEQ ID NO: 1) to a child (e.g., a child of about 5 to about 12 years of age having HPP) that exhibited an average Peabody Developmental Motor Scales, 2nd Edition (PDMS-2) standard score indicative of delayed motor development (e.g., an average PDMS-2 standard score of about 5) at about 3 years of age or less than 3 years of age are described.

Methods for administering asfotase alfa (SEQ ID NO: 1) to a child (e.g., a child of about 5 to about 12 years of age having HPP) having one or more (e.g., two, three, four, or five) scores selected from the average BOT-2 score, average CHAQ index score, average PODCI score, average 6MWT distance, average BSID-III score, and average PDMS-2 standard score are further described. Alternatively, the child (e.g., a child of about 5 to about 12 years of age having HPP) may not have an average BOT-2 score, average CHAQ index score, average PODCI score, average 6MWT distance, average BSID-III score, or average PDMS-2 standard score, and instead exhibits symptoms of physical impairments, pain, and/or development delay as described. Moreover, the child may have a 6MWT distance that is indicative of impairments in walking ability.

In any of these methods, asfotase alfa (SEQ ID NO: 1) may be administered to the child (e.g., a child of about 5 to about 12 years of age having HPP) for an extended period of time, e.g., at least four years, at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient). Furthermore, given the results described herein using asfotase alfa, other sALPs (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) are useful for treating a child having HPP (e.g., a child of about 5 to about 12 years of age having HPP) for an extended period of time, e.g., at least four years, at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient) as described.

Methods of Treatment

Provided herein are methods for treating a child having HPP (e.g., a child of about 5 to about 12 years of age having HPP). Children having HPP can be treated by administering a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) across a range of ages, e.g., about 5 to about 12 years of age, about 6 to about 7 years of age, about 6 to about 8 years of age, about 7 to about 9 years of age, about 8 to about 10 years of age, about 8 to about 9 years of age, about 9 to about 11 years of age, about 10 to about 11 years of age, about 10 to about 12 years of age, about 11 to about 12 years of age, about 5 to about 10 years of age, about 5 to about 8 years of age, about 5 to about 6 years of age, or about 5 to about 11 years of age.

Children (e.g., children of about 5 to about 12 years of age) can be diagnosed with HPP prior to administration of a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa). A child having HPP (e.g., a child of about 5 to about 12 years of age having HPP) can exhibit, e.g., physical impairments, disability in ADL, pain, and/or delayed motor development, relative to a child without HPP (e.g., a child of about 5 to about 12 years of age without HPP). Additionally, the child having HPP (e.g., a child of about 5 to about 12 years of age having HPP) can be a naïve patient that has not have previously received treatment with a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa).

The method involves administering a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) to a child having HPP (e.g., a child of about 5 to about 12 years of age having HPP), such as administering a sALP for a period of least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years). In particular, a sALP, such as asfotase alfa, can be administered to a child (e.g., a child of about 5 to about 12 years of age) previously determined to have an average BOT-2 strength score of less than 10, an average BOT-2 running speed and agility score of less than 5, an average CHAQ index score greater than about 0.8, an average PODCI score of less than about 40, and/or an average 6MWT distance less than 80% of the predicted 6MWT distance for a period of least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years).

Alternatively, the sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can be administered to the patient having HPP (e.g., a child, particularly a child of about 5 to about 12 years of age or an infant, particularly an infant of about 3 years of age or less than 3 years of age such as 6 months, 1 year of age, and 2-years of age) prior to determination of such scores (e.g., the BOT-2 strength score, BOT-2 running speed and agility score, the CHAQ index score, a 6MWT distance, the BSID-III scaled score, and/or the PDMS-2 standard score) to allow for, e.g., an increase in activities of ADL, a decrease in pain, and/or improved motor development.

Additionally, each of the described scores (e.g., the BOT-2 strength score, BOT-2 running speed and agility score, the CHAQ index score, the 6MWT distance, the BSID-III scaled score, and/or the PDMS-2 standard score) of a patient having HPP (e.g., a child of about 5 to about 12 years of age having HPP) can be used singly or in combination to assess treatment efficacy using a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), in which improvements relative to a certain test score demonstrate that the sALP is effective for treating HPP. For example, when administration of a sALP to a child having HPP (e.g., a child of about 5 to about 12 years of age) results in an average increase in the BOT-2 strength score to about 10 or greater than about 10, in which the child previously had an average BOT-2 strength score of less than about 10, then the sALP treatment is effective at treating, e.g., physical impairments associated with HPP. Alternatively, when administration of a sALP does not result in an average increase in the BOT-2 strength score to about 10 or greater than about 10, the dosage and/or frequency of sALP administration can be changed in order to determine the effective amount of the sALP for the child having HPP (e.g., a child of about 5 to about 12 years of age). For instance, the dosage of the sALP such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can be increased from, e.g., about 3 mg/kg/week to about 6 mg/kg/week or about 6 mg/kg/week to about 9 mg/kg/wk.

Hypophosphatasia in Children

Asfotase alfa is administered, as described herein, to treat, e.g., perinatal HPP, infantile HPP, childhood HPP, and odonto-HPP. In particular, patients having childhood HPP (e.g., children of about 5 to about 12 years of age having HPP) or infantile HPP (e.g., infants of about 3 years of age or less than 3 years of age) can be treated with a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) for a period of at least one year (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., the lifetime of the patient)).

In preferred embodiments, an HPP phenotype, e.g., perinatal HPP, infantile HPP, childhood HPP, and odontohypophosphatasia, is treated with a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa). For instance, the methods are useful for treating a perinatal HPP patient, such as a patient with increased respiratory compromise due to hypoplastic and rachitic disease of the chest; diminished ossification of the skull; diminished ossification and height of vertebral bodies; and/or absent ossification of the humeral, radial, and ulnar metaphyses with marked metaphyseal irregularity; fragmentation and fraying. The methods are also useful for treating patients exhibiting symptoms of infantile HPP, including, but not limited to, inadequate weight gain, the appearance of rickets, impaired skeletal mineralization, progressive skeletal demineralization, rib fractures, and chest deformity. A patient with childhood HPP, such as patients exhibiting symptoms such as premature loss of deciduous teeth (e.g., as a result of aplasia, hypoplasia, or dysplasia of dental cementum) and rickets, which causes short stature and skeletal deformities, such as bowed legs and enlarged wrists, knees, and ankles as a result of flared metaphyses, are treated with the methods described herein.

Accordingly, the methods are useful for alleviating any of the symptoms of HPP described herein, particularly when the sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) is administered for a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)). Non-limiting examples of HPP symptoms that can be treated with a sALP for a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)) include elevated blood and/or urine levels of inorganic pyrophosphate (PR), elevated blood and/or urine levels of phosphoethanolamine (PEA), elevated blood and/or urine levels of pyridoxal 5′-phosphate (PLP), hypomineralization, rachitic ribs, hypercalciuria, bone pain, bone fracture, HPP-related seizure, inadequate weight gain, and/or calcium pyrophosphate dihydrate crystal deposition.

A patient (e.g., a child, particularly a child of about 5 to about 12 years of age, or an infant, particularly an infant of about 3 years of age or less than 3 years of age) with a mutation in TNALP can also be treated with a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), e.g., to alleviate pain, increase ADL, and/or improve motor development. Missense mutations at a variety of positions in TNALP, including the enzyme's active site vicinity, homodimer interface, crown domain, amino-terminal arm, and calcium-binding site, have all been found to affect its catalytic activity. In addition, missense, nonsense, frame-shift, and splice site mutations have also been shown to lead to aberrant mutant proteins or intracellular trafficking defects that lead to subnormal activity on the cell surface. Accordingly, the methods can be used to treat patients with different mutation in TNALP (e.g., missense mutations, frame-shift, nonsense, and splicing mutations).

For instance, the presence of a mutation in TNALP can be detected in a sample from the patient (e.g., a child, particularly a child of about 5 to about 12 years of age, or an infant, particularly an infant of about 3 years of age or less than 3 years of age) prior to or after treatment (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa). Additionally, a parent of the patient and/or a fetal sample (e.g., fetal nucleic acid obtained from maternal blood, placental, and/or fetal samples) can be tested by methods known in the art for a mutation in TNALP. Traditional management of HPP has also included symptomatic treatment of the phenotypic manifestations of the disease, e.g., treating hypercalcemia with dietary restriction or calciuretics and orthopedic stabilization of fractures. Accordingly, these treatments (e.g., dietary restriction, calciuretics, and orthopedic stabilization of fractures) can be used with a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) administered for a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)), such as to decrease physical impairments, increase ADL, alleviate pain, and/or improve motor development.

Exemplary tests useful in the methods include (1) the Bruininks-Oseretsky Test of Motor Proficiency 2nd Edition (BOT-2), (2) the Childhood Health Assessment Questionnaire (CHAQ), (3) the Pediatric Outcomes Data Collection Instrument (PODCI), (4) the Six Minute Walk Test (6MWT), (5) the Bayley Scales of Infant and Toddler Development, 3rd Edition (BSID-III), and (6) the Peabody Developmental Motor Scales, 2nd Edition (PDMS-2), which are described in further detail below.

Bruininks-Oseretsky Test of Motor Proficiency 2nd Edition (BOT-2)

Children with HPP (e.g., children of about 5 to about 12 years of age) can be identified for treatment with a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) using the Bruininks-Oseretsky Test of Motor Proficiency 2nd Edition (BOT-2), which is described in Bruininks, R. H. (2005). Bruininks-Oseretsky Test of Motor Proficiency, (BOT-2), Minneapolis, Minn.: Pearson Assessment, hereby incorporated by reference in its entirety. In particular, the BOT-2 can be used to evaluate physical impairments and mobility restrictions in a child having HPP (e.g., children of about 5 to about 12 years of age having HPP) to generate a BOT-2 score for the child.

The BOT-2 includes a range of tests to evaluate physical impairments of a child (e.g., a child of about 5 to about 12 years of age), which can be performed with, e.g., a kit including the tests. The BOT-2 provides composite BOT-2 scores in the following areas: strength, running speed and agility, fine motor precision, fine motor integration, manual dexterity, bilateral coordination, balance, and upper-limb coordination. For example, the child having HPP (e.g., a child of about 5 to about 12 years of age having HPP) can perform sit-ups, v-ups, standing long jump, wall sit, and/or push-ups to determine the BOT-2 strength score; the child having HPP (e.g., a child of about 5 to about 12 years of age having HPP) can step over a balance beam and/or perform a shuttle run, two-legged side hop, and/or one-legged side hop to determine the BOT-2 running speed and agility score; the child having HPP (e.g., a child of about 5 to about 12 years of age having HPP) can cut out a circle and/or connect dots to determine the BOT-2 fine motor precision score; the child having HPP (e.g., a child of about 5 to about 12 years of age having HPP) can copy a star and/or copy a square to determine the BOT-2 fine motor integration score; the child having HPP (e.g., a child of about 5 to about 12 years of age having HPP) can transfer pennies, sort cards, and/or string blocks to determine the manual dexterity score; the child having HPP (e.g., a child of about 5 to about 12 years of age having HPP) can tap his or her foot and finger and/or perform jumping jacks to determine the BOT-2 bilateral coordination score; the child having HPP (e.g., a child of about 5 to about 12 years of age having HPP) can walk forward on a line and/or stand on one leg on a balance beam to determine the BOT-2 balance score; and the child having HPP (e.g., a child of about 5 to about 12 years of age having HPP) can throw a ball at a target and/or catch a tossed ball to determine the BOT-2 upper-limb coordination score.

A child having HPP (e.g., a child of about 5 to about 12 years of age having HPP) could perform tests in one or more of described areas (strength, running speed and agility, fine motor precision, fine motor integration, manual dexterity, bilateral coordination, balance, and upper-limb coordination) to generate a BOT-2 score indicative of physical impairments in the child. Within each BOT-2 area (strength, running speed and agility, fine motor precision, fine motor integration, manual dexterity, bilateral coordination, balance, and upper-limb coordination), a child having HPP could perform one or more tests to determine the BOT-2 score of the child, e.g., the child could perform one or more of sit-ups, v-ups, standing long jump, wall sit, and push-ups to determine the BOT-2 strength score. Thus, only one test (e.g., one test selected from the group of sit-ups, v-ups, standing long jump, wall sit, and push-ups) can be performed to determine the BOT-2 score (e.g., a BOT-2 strength score) of a child having HPP.

Each of the BOT-2 scores (strength, running speed and agility, fine motor precision, fine motor integration, manual dexterity, bilateral coordination, balance, and upper-limb coordination) of the patient having HPP (e.g., a child of about 5 to about 12 years of age having HPP) can be compared to the BOT-2 score of patients without HPP (e.g., children of about 5 to about 12 years of age without HPP) to, e.g., determine the standard deviation of the BOT-2 score. Each of the BOT-2 scores (e.g., strength, running speed and agility, fine motor precision, fine motor integration, manual dexterity, bilateral coordination, balance, and upper-limb coordination) of the patient having HPP (e.g., a child of about 5 to about 12 years of age having HPP) can be compared to the BOT-2 score of other HPP patients (e.g., children of about 5 to about 12 years of age having HPP) to, e.g., determine the average BOT-2 score for the HPP patient.

BOT-2 scores (e.g., strength, running speed and agility, fine motor precision, fine motor integration, manual dexterity, bilateral coordination, balance, and upper-limb coordination scores) range from about 0 to equal to or less than about 25, in which a score of about 10 to about 20 is considered representative of healthy patients (e.g., patients without HPP). Patients with an average BOT-2 score (e.g., strength, running speed and agility, fine motor precision, fine motor integration, manual dexterity, bilateral coordination, balance, and upper-limb coordination scores) of less than about 10 can be treated with a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), such as by administering a sALP for a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)).

For example, HPP patients (e.g., a child of about 5 to about 12 years of age having HPP) with a BOT-2 strength score of less than 10 (e.g, about 0, about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, or about 10) can be treated with a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) for a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)). Likewise, HPP patients (e.g., a child of about 5 to about 12 years of age having HPP) with a BOT-2 running speed and agility score of less than 10 (e.g, about 0, about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, or about 10) can then be treated with a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) for a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)).

The methods can result in an improvement in the BOT-2 score (e.g., strength, running speed and agility, fine motor precision, fine motor integration, manual dexterity, bilateral coordination, balance, and/or upper-limb coordination score) of a HPP patient (e.g., a child of about 5 to about 12 years of age having HPP). For example, treatment with a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), such as treatment with a sALP for a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)), can result in an average increase in the BOT-2 strength score to about 10 to about 20 (e.g. about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, or about 20). Additionally, treatment with a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), such as treatment with a sALP for a period of a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)), can result in an average increase in the BOT-2 running speed and agility score to about 5 to about 20 (e.g. about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, or about 20).

The increase in the BOT-2 score (e.g., strength, running speed and agility, fine motor precision, fine motor integration, manual dexterity, bilateral coordination, balance, and/or upper-limb coordination score) can be sustained throughout administration of the sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), e.g., for a period of a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)). Likewise, the decrease in physical impairments after administration of the sALP can be sustained throughout administration of the sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), e.g., a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)).

The BOT-2 scores (strength, running speed and agility, fine motor precision, fine motor integration, manual dexterity, bilateral coordination, balance, and upper-limb coordination scores) of a patient having HPP (e.g., a child of about 5 to about 12 years of age having HPP) can be used singly or in combination to assess treatment efficacy using a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), in which improvements relative to a certain test score demonstrate that the sALP is effective for treating physical impairments associated with HPP. For example, when administration of a sALP to a child having HPP (e.g., a child of about 5 to about 12 years of age) results in an average increase in the BOT-2 running speed and agility score to about 5 or greater than about 5, in which the child previously had an average BOT-2 running speed and agility score of less than about 5, then the sALP is considered to be effective at, e.g., treating physical impairments associated with HPP.

Additionally, within each BOT-2 area (strength, running speed and agility, fine motor precision, fine motor integration, manual dexterity, bilateral coordination, balance, and upper-limb coordination), a child having HPP could perform one or more tests to determine the BOT-2 score of the child. For instance, the child having HPP could perform one or more of sit-ups, v-ups, standing long jump, wall sit, and push-ups to determine the BOT-2 strength score, to determine the BOT-2 strength score and assess the treatment efficacy of sALP administration. The child having HPP could perform one or more of balance beam, a shuttle run, two-legged side hop, and/or one-legged side hop to determine the BOT-2 running speed and agility score and assess the treatment efficacy of sALP administration. The child having HPP can cut out a circle and/or connect dots to determine the BOT-2 fine motor precision score and assess the treatment efficacy of sALP administration. The child having HPP can copy a star and/or copy a square to determine the BOT-2 fine motor integration score and assess the treatment efficacy of sALP administration. The child having HPP could perform one or more of transferring pennies, sorting cards, and stringing blocks to determine the BOT-2 manual dexterity score and assess the treatment efficacy of sALP administration. The child having HPP can tap his or her foot and finger and/or perform jumping jacks to determine the BOT-2 bilateral coordination score and assess the treatment efficacy of sALP administration. The child having HPP can walk forward on a line and/or stand on one leg on a balance beam to determine the BOT-2 balance score and assess the treatment efficacy of sALP administration. The child having HPP can throw a ball at a target and/or catch a tossed ball to determine the BOT-2 upper-limb coordination score and assess the treatment efficacy of sALP administration.

Alternatively, when administration of a sALP does not result in an average increase in the BOT-2 running speed and agility score to greater than about 5, the dosage and/or frequency of sALP administration can be changed in order to determine the effective amount of the sALP for the child having HPP (e.g., a child of about 5 to about 12 years of age). For instance, the dosage of the sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can be increased from, e.g., about 3 mg/kg/week to about 6 mg/kg/week or about 6 mg/kg/week to about 9 mg/kg/wk.

Childhood Health Assessment Questionnaire (CHAQ)

Children with HPP (e.g., children of about 5 to about 12 years of age) can be identified for treatment with a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) using the Childhood Health Assessment Questionnaire (CHAQ). The CHAQ can be administered to evaluate the health status of children having HPP (e.g., children of about 5 to about 12 years of age having HPP) to generate a CHAQ index score for the child, as is described in Bruce & Fries (J. Rheumatol. 30(1): 167-178, 2003) and Klepper (Arthritis & Rheumatism, 49: S5-S14, 2003), hereby incorporated by reference in their entirety. The CHAQ includes eight categories of questions for dressing/grooming, arising, eating, walking, hygiene, reach, grip, and activities, in which a parent or guardian records the amount of difficulty the child with HPP (e.g., a child of about 5 to about 12 years of age having HPP) has in performing the respective activities. The range of scores within each category is from 0 to 3, in which a score of 0 indicates without any difficulty; a score of 1 indicates with some difficulty; a score of 2 indicates with much difficulty; and a score of 3 indicates that the child is unable to perform the activity.

Children with HPP (e.g., children of about 5 to about 12 years of age) with an average CHAQ index score (e.g., indicative of disability in activities of daily living (ADL) and/or pain) greater than about 0.8 (e.g., about 0.8, about 1, about 1.2, about 1.4, about 1.6, about 1.8, about 2.0, about 2.2, about 2.4, about 2.6, about 2.8, or about 3.0) can be treated by administering a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa). In particular, children with an average CHAQ index score of greater than about 0.8 can be treated by administering a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) for a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the children)). Furthermore, a child having HPP could be asked one or more questions in one or more of the eight categories (dressing/grooming, arising, eating, walking, hygiene, reach, grip, and activities) to arrive at an average CHAQ index score, and if the average CHAQ index score is greater than about 0.8, the child can be treated by administering a sALP.

The CHAQ index score of a child having HPP (e.g., a child of about 5 to about 12 years of age having HPP) can be compared to the CHAQ index score of children without HPP (e.g., children of about 5 to about 12 years of age without HPP) to, e.g., determine the standard deviation of the CHAQ index score. Additionally, the CHAQ index score of a child having HPP (e.g., a child of about 5 to about 12 years of age having HPP) can be compared to the CHAQ index score of other children having HPP (e.g., children of about 5 to about 12 years of age without HPP) to, e.g., determine the standard deviation of the CHAQ index score.

The methods can result in an improvement in the CHAQ index score (e.g., indicative of disability in ADL and/or pain) of the child having HPP (e.g., a child of about 5 to about 12 years of age having HPP). For example, treatment with a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), such as treatment with a sALP for a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)) can result in an average decrease in the CHAQ index score to about 0 to equal to or less than about 0.5 (e.g. about 0, about 0.1, about 0.2, about 0.4, or about 0.5).

The increase in the CHAQ index score of the child having HPP (e.g., a child of about 5 to about 12 years of age having HPP) can be sustained throughout administration of the sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), e.g., for a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)). Likewise, the increase in ADL and/or decrease in pain of the child having HPP (e.g., a child of about 5 to about 12 years of age having HPP) can be sustained throughout administration of the sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), e.g., a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)).

The CHAQ index score of a child having HPP (e.g., a child of about 5 to about 12 years of age having HPP) can be used to assess treatment efficacy using a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), in which improvements relative to a certain test score demonstrate that the sALP is effective for treating, e.g., disability in activities of daily living (ADL) and pain associated with HPP. In particular, a child having HPP could be asked one or more questions in one or more of the eight categories (dressing/grooming, arising, eating, walking, hygiene, reach, grip, and activities) to arrive at an average CHAQ index score and to assess treatment efficacy of sALP administration. For example, when administration of a sALP to a child having HPP (e.g., a child of about 5 to about 12 years of age) results in an average decrease in the CHAQ index score to equal to or less than about 0.5, in which the child previously had an average CHAQ index score of greater than about 0.8, then the sALP is effective at treating, e.g., disability in activities of daily living (ADL) and pain associated with HPP. Alternatively, when administration of a sALP does not result in an average decrease in the CHAQ index score to equal to or less than about 0.5, the dosage and/or frequency of sALP administration can be changed in order to determine the effective amount of the sALP for the child having HPP (e.g., a child of about 5 to about 12 years of age). For instance, the dosage of the sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can be increased from, e.g., about 3 mg/kg/week to about 6 mg/kg/week or about 6 mg/kg/week to about 9 mg/kg/wk.

Pediatric Outcomes Data Collection Instrument (PODCI)

Certain patients with HPP (e.g., children of about 5 to about 12 years of age having HPP) can be identified for treatment with a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) using the Pediatric Outcomes Data Collection Instrument (PODCI). The PODCI can be administered to evaluate the health status of children having HPP under about 19 years of age (e.g., children of about 5 to about 12 years of age having HPP) to generate a PODCI score for the patient, as is described in Plint et al. (J. Pediatr. Orthop. 23(6): 788-790, 2003). The PODCI includes eight categories of questions that can be completed by the patient or by a parent/guardian of the HPP patient. Categories that can be used to determine the PODCI of a HPP patient (e.g., a child of about 5 to about 12 years of age having HPP) include the following: 1) the upper extremity and physical function scale to measure difficulty encountered in performing daily personal care and student activities; 2) the transfer and basic mobility scale to measure difficulty experienced in performing routine motion and motor activities in daily activities; 3) the sports/physical functioning scale to measure difficulty or limitations encountered in participating in more active activities or sports; 4) the pain/comfort scale to measure the level of pain experienced during the past week; 5) the treatment expectations scale to measure the long term expectations of treatment; 6) the happiness scale to measure overall satisfaction with personal looks and sense of similarity to friends and others of own age; 7) the satisfaction with symptoms scale to measure the patient's acceptance of current limitations should this be a life-long state; and 8) the global functioning scale, which is a general combined scale calculated from the first four scales listed above. In each of the categories, a standardized score is determined for the HPP patient (e.g., a child of about 5 to about 12 years of age having HPP) and then converted to a 0 to 100 scale, in which 0 represents significant disability and 100 represents less disability.

HPP patients (e.g., children of about 5 to about 12 years of age having HPP) with an average PODCI score (e.g., indicative of disability in ADL and/or pain) less than about 40 (e.g., about 5, about 10, about 15, about 20, about 25, about 30, about 35, or about 39) can be treated by administering a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa). In particular, patients (e.g., children of about 5 to about 12 years of age having HPP) with an average PODCI score of less than 40 can be treated by administering a sALP for a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)). Furthermore, a child having HPP could be asked one or more questions in one or more of the eight scales described above (e.g., transfer and basic mobility, sports/physical functioning, and the pain/comfort scale) to arrive at an average PODCI score, and if the average PODCI score is greater than less than 40, the child can be treated by administering a sALP.

The methods described herein can result in an increase in the PODCI score (e.g., indicative of disability in ADL and/or pain) of the HPP patient (e.g., children of about 5 to about 12 years of age having HPP). For example, treatment with a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), such as treatment with a sALP for a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)), can result in an average increase in the PODCI score to about 40 to about 50 (e.g. about 40, about 41, about 42, about 43, about 44, about 45, about 46, about 47, about 48, about 49, or about 50).

The increase in the PODCI score can be sustained throughout administration of the sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), e.g., for a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)). Likewise, the increase in ADL and/or decrease in pain can be sustained throughout administration of the sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), e.g., a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)).

The PODCI score of a patient having HPP (e.g., a child of about 5 to about 12 years of age having HPP) can be used to assess treatment efficacy using a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), in which improvements relative to a certain test score demonstrate that the sALP is effective for treating, e.g., disability in activities of daily living (ADL) and pain associated with HPP. In particular, a child having HPP could be asked one or more questions in one or more of the eight scales (the upper extremity and physical function scale, the transfer and basic mobility scale, the sports/physical functioning scale, the pain/comfort scale, the treatment expectations scale, the happiness scale, the satisfaction with symptoms scale, and the global functioning scale) to arrive at an average PODCI score and to assess treatment efficacy of sALP administration.

For example, when administration of a sALP to a child having HPP (e.g., a child of about 5 to about 12 years of age) results in an average increase in the PODCI score to about 40 or greater than about 40, in which the child previously had an average PODCI score of less than about 40, then the sALP is effective at treating, e.g., disability in activities of daily living (ADL) and pain associated with HPP. Alternatively, when administration of a sALP does not result in an average increase in the PODCI score to about 40 or greater than about 40, the dosage and frequency of sALP administration can be changed in order to determine the effective amount of the sALP for the child having HPP (e.g., a child of about 5 to about 12 years of age). For instance, the dosage of the sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can be increased from, e.g., about 3 mg/kg/week to about 6 mg/kg/week or about 6 mg/kg/week to about 9 mg/kg/wk.

Six Minute Walk Test (6MWT)

Children with HPP (e.g., children with HPP of about 5 to about 12 years of age) can be identified for treatment with a sALP (such as TNALP, for example the sALP fusion polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) using the 6MWT. In particular, the 6MWT can be used to evaluate walking ability in children with HPP (e.g., children with HPP of about 5 to about 12 years of age) to determine the 6MWT distance for the child prior to or after administration of a sALP.

The 6MWT can be performed indoors or outdoors using a flat, straight, enclosed corridor (e.g., of about 30 meters in length) with a hard surface. A stopwatch or other timer can be used to track the time and a mechanical counter or other device can be used to determine the distance (e.g., in meters) that the HPP patient (e.g., a child with HPP of about 5 to about 12 years of age) walks. For instance, the length of the corridor can be marked every three meters to determine the number of meters walked by the HPP patient, with the turnaround point at 30 meters and the starting line also marked. The distance walked by the patient in six minutes (the 6MWT distance) can then be compared to the predicted number of meters walked, e.g., by an untreated or healthy subject of about the same age, the same gender, and/or the same height, and expressed as a percentage value to generate the 6MWT value of the patient. The 6MWT distance of the patient (e.g., the child having HPP) can also be compared to the 6MWT distance of the patient at baseline.

HPP patients (e.g., a child with HPP of about 5 to about 12 years of age) with an average 6MWT distance of less than about 80% of the predicted 6MWT distance can be treated with a sALP (such as TNALP, for example the sALP fusion polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), such as by administering a sALP for a period of at least one year (e.g., at least two years, at least three years, at least four years, at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or longer than ten years, such as for the lifetime of the patient). For example, an HPP patient with an average 6MWT of less than about 80% of the predicted 6MWT distance (e.g., about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, or about 75% of the predicted 6MWT distance) can be treated with a sALP for a period of at least one year (e.g., at least two years, at least three years, at least four years, at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or longer than ten years, such as for the lifetime of the patient).

The methods can result in an improvement in the 6MWT distance of a HPP patient (e.g., a child with HPP of about 5 to about 12 years of age). For example, treatment with a sALP (such as TNALP, for example the sALP fusion polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), such as treatment with a sALP for a period of at least one year (e.g., at least two years, at least three years, at least four years, at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or longer than ten years, such as for the lifetime of the patient), can result in an average increase in the 6MWT distance of about 20 meters or more (e.g., about 20 meters, about 25 meters, about 30 meters, about 35 meters, about 40 meters, about 45 meters, about 50 meters, about 55 meters, about 60 meters, about 65 meters, about 70 meters, about 75 meters, about 80 meters, about 85 meters, about 90 meters, about 95 meters, about 100 meters, or more). Likewise, treatment with a sALP can result in an average increase in the 6MWT value to about 50% or greater of the predicted 6MWT value (e.g., the predicted number of meters walked by a healthy subject of about the same age, the same gender, and/or the same height expressed as a percentage value) of the patient (e.g. about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or more of the predicted 6MWT value).

The increase in the 6MWT distance of the HPP patient (e.g., a child with HPP of about 5 to about 12 years of age) can be sustained throughout administration of the sALP (such as TNALP, for example the sALP fusion polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), e.g., for a period of at least one year (e.g., at least two years, at least three years, at least four years, at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or longer than ten years, such as for the lifetime of the patient). For instance, the 6MWT value increases by about 20 meters and/or to greater than about 80% of the predicted 6MWT distance of the patient and remains at ±10% of the increased 6MWT distance during treatment with the sALP (e.g., asfotase alfa).

Likewise, the improvement in walking ability of the HPP patient can be sustained throughout administration of the sALP, e.g., for a period of at least one year (e.g., at least two years, at least three years, at least four years, at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or longer than ten years, such as for the lifetime of the patient). For instance, the HPP patient exhibits decreased reliance on an assistive device for walking, such as a wheelchair, a wheeled walker, a cane, or an orthotic during treatment with the sALP.

Alternatively, when administration of a sALP does not result in an average increase in the 6MWT distance by about 20 meters relative to baseline, the dosage and/or frequency of sALP administration can be changed in order to determine the effective amount of the sALP for the HPP patient (e.g., a child with HPP of about 5 to about 12 years of age). Likewise, when administration of a sALP does not result in an average increase in the 6MWT distance to greater than 80% of the predicted 6MWT distance (e.g., of an untreated subject having HPP of about the same age, same gender, and/or height), the dosage and/or frequency of sALP administration can be changed in order to determine the effective amount of the sALP for the HPP patient (e.g., a child with HPP of about 5 to about 12 years of age). For instance, the dosage of the sALP (such as TNALP, for example the sALP fusion polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can be increased from, e.g., about 2.1 mg/kg/week or about 3.5 mg/kg/week to about 6 mg/kg/week or about 9 mg/kg/week.

Bayley Scales of Infant and Toddler Development, 3rd Edition (BSID-III)

Children with HPP (e.g., children of about 5 to about 12 years of age) can be identified for treatment with a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) using the Bayley Scales of Infant and Toddler Development, 3rd Edition (BSID-III). The BSID-III can be administered to evaluate the health status of HPP patients from birth to generate a BSID-III score for the patient, as is described in Bayley. (2006). Bayley scales of infant and toddler development: administration manual. San Antonio, Tex.: Harcourt Assessment. The BSID-III includes a series of developmental play tasks that can be administered to the HPP patient (e.g., infants of about three years of age or less having HPP) to determine the raw BSID-III score. For example, categories for determining the BSID-III score of an HPP patient (e.g., infants of about three years of age or less having HPP) can include prehension, perceptual-motor integration, motor planning and speed, visual tracking, reaching, object grasping, object manipulation, functional hand skills, responses to tactile information, movement of the limbs and torso, static positioning, dynamic movement, balance, and motor planning. The BSID-III measurements are then converted to scaled BSID-III scores, which can be used to determine the HPP patient's performance compared to healthy, age-adjusted patients. The BSID-III scaled score of a patient (e.g., a patient with HPP) can range from 0 to 14, in which scores of about 7 to about 13 are considered the normal range of healthy patients.

A child having HPP (e.g., a child of about 5 to about 12 years of age having HPP) could perform tests in one or more of described categories (prehension, perceptual-motor integration, motor planning and speed, visual tracking, reaching, object grasping, object manipulation, functional hand skills, responses to tactile information, movement of the limbs and torso, static positioning, dynamic movement, balance, and motor planning) as an infant (e.g., at about 3 years of age or less than 3 years of age) to generate a BSID-III score indicative of delayed motor development. Children having HPP with an average BSID-III score in one or more of the described categories (prehension, perceptual-motor integration, motor planning and speed, visual tracking, reaching, object grasping, object manipulation, functional hand skills, responses to tactile information, movement of the limbs and torso, static positioning, dynamic movement, balance, and motor planning) less than about 2 as an infant can be treated by administering a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa). In particular, children having HPP with an average BSID-III score of less than about 2 as an infant can be treated by administering a sALP for a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)).

The methods can result in an improvement in the average BSID-III score (e.g., indicative of delayed motor development) of the HPP patient. For example, treatment with a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), such as treatment with a sALP for a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)) can result in an average increase in the BSID-III score to greater than about 5 (e.g., about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, or about 13).

The increase in the BSID-III score can be sustained throughout administration of the sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), e.g., for a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)). Likewise, the increase in motor development can be sustained throughout administration of the sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), e.g., for a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)).

The BSID-III score of a patient having HPP (e.g., a child of about 5 to about 12 years of age having HPP) can be used to assess treatment efficacy using a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), in which improvements relative to a certain test score demonstrate that the sALP is effective for treating, e.g., delayed motor development associated with HPP. In particular, a child having HPP could perform tests in one or more of described categories (prehension, perceptual-motor integration, motor planning and speed, visual tracking, reaching, object grasping, object manipulation, functional hand skills, responses to tactile information, movement of the limbs and torso, static positioning, dynamic movement, balance, and motor planning) as an infant (e.g., at about three years of age or less having HPP) to arrive at an average BSID-III score and to assess treatment efficacy of sALP administration.

For example, when administration of a sALP to a child having HPP (e.g., a child of about 5 to about 12 years of age) results in an average increase in the BSID-III scaled score to greater than about 5, in which the child previously had an average BSID-III scaled score of less than about 2 as an infant (e.g., at about 3 years of age or less than 3 years of age), then the sALP is effective at treating, e.g., delayed motor development associated with HPP. Alternatively, when administration of a sALP does not result in an average increase in the BSID-III scaled score to greater than about 5, the dosage and/or frequency of sALP administration can be changed in order to determine the effective amount of the sALP for the child having HPP (e.g., a child of about 5 to about 12 years of age). For instance, the dosage of the sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can be increased from, e.g., about 3 mg/kg/week to about 6 mg/kg/week or about 6 mg/kg/week to about 9 mg/kg/wk.

Peabody Developmental Motor Scales, 2nd Edition (PDMS-2)

Children with HPP (e.g., children of about 5 to about 12 years of age) can be identified for treatment with a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) using the Peabody Developmental Motor Scales, 2nd Edition (PDMS-2). The PDMS-2 can be administered to evaluate the health status of HPP patients from birth to generate a PDMS-2 score for the patient, as is described in van Hartingsveldt, et al. (Occup. Ther. Int. 12(1): 1-13, 2005). The PDMS-2 includes six categories of subtests to measure motor skills of patients, such as a patient having HPP (e.g., infants and children having HPP).

In particular, PDMS-2 measurements can be determined from the following subtests: 1) the locomotor subtest to measure a child's (e.g., a child having HPP of about 5 years of age or less than 5 years of age) ability to move from one place to another (measurements include crawling, walking, running, hopping, and jumping forward); 2) the reflexes subtest to measure a child's (e.g., a child having HPP of about 5 years of age or less than 5 years of age) ability to automatically react to environmental events; 3) the stationary subtest to measure a child's (e.g., a child having HPP of about 5 years of age or less than 5 years of age) ability to sustain control of his or her body within the center of gravity and retain equilibrium; 4) the object manipulation subtest to measure a child's (e.g., a child having HPP of about 5 years of age or less than 5 years of age) ability to manipulate an object, such as catching, throwing, and kicking a ball; 5) the grasping subtest to measure a child's (e.g., a child having HPP of about 5 years of age or less than 5 years of age) ability to use his or her hands, such as the ability to hold an object with one hand and actions involving the controlled use of the fingers of both hands; and 6) the visual-motor integration subtest to measure a child's (e.g., a child having HPP of about 5 years of age or less than 5 years of age) ability to use his or her visual perceptual skills to perform complex eye-hand coordination tasks, such as reaching and grasping for an object, building with blocks, and copying designs. The PDMS-2 measurement can be determined for one or more of these categories for children with HPP (e.g., children of about 5 years of age or less than 5 years of age) and then converted into a PDMS-2 score, such as the PDMS-2 locomotor standard score ranging from 0 to 13, in which the range of healthy patients (e.g., patients without HPP) is from about 7 to about 13.

Children with HPP (e.g., children of about 5 to about 12 years of age) with an average PDMS-score (e.g., indicative of delayed motor development) of about 5 at 5 years of age or less than 5 years of age can be treated by administering a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa). In particular, children having HPP with an average PDMS-score in one or more of the described categories (locomotor, reflexes, stationary, object manipulation, grasping, and visual-motor) of about 5 at 5 years of age or less than 5 years of age can be treated by administering a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa).

The methods described herein can result in an improvement in the PDMS-2 score (e.g., indicative of delayed motor development) of the HPP patient (e.g., children of about 5 to about 12 years of age). For example, treatment with a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), such as treatment with a sALP for a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)) can result in an average increase in the PDMS-2 score to about 7 to about 13 (e.g., about 7, about 8, about 9, about 10, about 11, about 12, or about 13).

The increase in the PDMS-2 score can be sustained throughout administration of the sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), e.g., for a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)). Likewise, the increase in motor development can be sustained throughout administration of the sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) for a period of at least four years (e.g., at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)).

The PDMS-2 score of a child having HPP (e.g., a child of about 5 to about 12 years of age having HPP) can be used to assess treatment efficacy using a sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa), in which improvements relative to a certain test score demonstrate that the sALP is effective for treating, e.g., delayed motor development associated with HPP. In particular, a child having HPP could perform tests in one or more of described categories (locomotor, reflexes, stationary, object manipulation, grasping, and visual-motor) at about 5 years of age or less than 5 years of age to arrive at an average PDMS-2 score and to assess treatment efficacy of sALP administration.

For example, when administration of a sALP to a child having HPP (e.g., a child of about 5 to about 12 years of age) results in an average increase in the PDMS-2 standard score to about 7, in which the child previously had an average PDMS-2 standard score of about 5, then the sALP is effective at treating, e.g., delayed motor development associated with HPP. Alternatively, when administration of a sALP does not result in an average increase in the PDMS-2 standard score to about 7, the dosage and/or frequency of sALP administration can be changed in order to determine the effective amount of the sALP for the child having HPP (e.g., a child of about 5 to about 12 years of age). For instance, the dosage of the sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can be increased from, e.g., about 3 mg/kg/week to about 6 mg/kg/week or about 6 mg/kg/week to about 9 mg/kg/wk.

Alkaline Phosphatase

Asfotase alfa is a human TNALP (hTNALP; SEQ ID NO: 1) fusion protein formulated for the treatment of HPP. In particular, asfotase alfa (SEQ ID NO: 1) can be used effectively to treat hypophosphatasia (HPP), its symptoms, and physical impairments associated therewith in a child with HPP (e.g., a child of about 5 to about 12 years of age having HPP) for an extended period of time (e.g., at least four years, at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years (e.g., for the lifetime of the patient)).

Given the results described herein, the present disclosure is not limited to a particular alkaline phosphatase (ALP) or nucleic acid sequence encoding an ALP. Alkaline phosphatases encompass a group of enzymes that catalyze the cleavage of a phosphate moiety (e.g., hydrolysis of pyrophosphate, PR). There are four known mammalian alkaline phosphatase (ALP) isozymes: tissue nonspecific alkaline phosphatase (TNALP; described further below), placental alkaline phosphatase (PLALP) (e.g., Accession Nos. P05187, NP_112603, and NP_001623), germ cell alkaline phosphatase (GALP) (e.g., Accession No. P10696), and intestinal alkaline phosphatase (IALP) (e.g., Accession Nos. P09923 and NP_001622). In addition to the exemplary ALPs discussed above, this disclosure also provides any polypeptide having the identical or similar catalytic site structure and/or enzymatic activity of ALP for treating HPP patients, such as children with HPP (e.g., children of about 5 to about 12 years of age having HPP) or infants with HPP (e.g., infants of about 3 years of age or less than 3 years of age). Bone delivery conjugates including sALP are further described in PCT publication Nos: WO 2005/103263 and WO 2008/138131.

TNALPs that can be used according to the methods described herein include, e.g., human TNALP (Accession Nos. NP_000469, AAI10910, AAH90861, AAH66116, AAH21289, and AAI26166); rhesus TNALP (Accession No. XP_01109717); rat TNALP (Accession No. NP_037191); dog TNALP (Accession No. AAF64516); pig TNALP (Accession No. AAN64273), mouse (Accession No. NP_031457), cow TNALP (Accession Nos. NP_789828, NP_776412, AAM 8209, and AAC33858), and cat TNALP (Accession No. NP_001036028). In particular, TNALP can be a recombinant human TNALP (e.g., SEQ ID NO: 1, asfotase alfa; see U.S. Pat. Nos. 7,763,712 and 7,960,529, incorporated herein by reference in their entirety) used for the treatment of HPP patients, such as children with HPP (e.g., children of about 5 to about 12 years of age having HPP) or infants with HPP (e.g., infants of about 3 years of age or less than 3 years of age). The TNALP can also be one that exhibits at least about 95% sequence identity to the polypeptide or nucleic acid sequence of the above-noted TNALPs.

Soluble Alkaline Phosphatase

The ALPs of the present invention include soluble (e.g., extracellular or non-membrane-bound) forms of any of the alkaline phosphatases described herein. The sALP of the invention can be, for example, a soluble form of human tissue non-specific alkaline phosphatase (human TNALP (hTNALP)). The present disclosure is not limited to a particular sALP and can include any sALP polypeptide that is physiologically active toward, e.g., phosphoethanolamine (PEA), inorganic pyrophosphate (PPi), and pyridoxal 5′-phosphate (PLP). In particular, a sALP of the present invention is catalytically competent to improve skeletal mineralization in bone. The present invention further includes nucleic acids encoding the sALPs described herein that can be used to treat the conditions described herein, e.g., HPP, such as children with HPP (e.g., children of about 5 to about 12 years of age having HPP) or infants with HPP (e.g., infants of about 3 years of age or less than 3 years of age).

TNALP is a membrane-bound protein anchored by a glycolipid moiety at the C-terminal (Swiss-Prot, P05186). This glycolipid anchor (GPI) is added post-translationally after the removal of a hydrophobic C-terminal end, which serves both as a temporary membrane anchor and as a signal for the addition of the GPI. While the GPI anchor is located in the cell membrane, the remaining portions of TNALP are extracellular. In particular, TNALP (e.g., human TNALP (hTNALP)) can be engineered to replace the first amino acid of the hydrophobic C-terminal sequence (an alanine) with a stop codon, thereby producing an engineered hTNALP that contains all amino acid residues of the native anchored form of TNALP and lacks the GPI membrane anchor. One skilled in the art will appreciate that the position of the GPI membrane anchor will vary in different ALPs and can include, e.g., the last 10, 12, 14, 16, 18, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 32, 34, 36, 38, 40, 45, 50, or more amino acid residues on the C-terminus of the polypeptide. Recombinant sTNALP can include, e.g., amino acids 1 to 502 (18 to 502 when secreted), amino acids 1 to 501 (18 to 501 when secreted), amino acids 1 to 504 (18 to 504 when secreted), amino acids 1 to 505 (18-505 when secreted), or amino acids 1 to 502. Thus, the C-terminal end of the native ALP can be truncated by certain amino acids without affecting ALP activity.

In addition to the C-terminal GPI anchor, TNALP also has an N-terminal signal peptide sequence. The N-terminal signal peptide is present on the synthesized protein when it is synthesized, but cleaved from TNALP after translocation into the ER. The sALPs of the invention include both secreted (i.e., lacking the N-terminal signal) and non-secreted (i.e., having the N-terminal signal) forms thereof. One skilled in the art will appreciate that the position of the N-terminal signal peptide will vary in different alkaline phosphatases and can include, for example, the first 5, 8, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 27, 30, or more amino acid residues on the N-terminus of the polypeptide. One of skill in the art can predict the position of a signal sequence cleavage site, e.g., by an appropriate computer algorithm such as that described in Bendtsen et al. (J. Mol. Biol. 340(4):783-795, 2004) and available on the Web at www.cbs.dtu.dk/services/SignalP/.

The present invention also includes sALP consensus sequences derived from the extracellular domain of ALP isozymes (e.g., TNALP, PALP, GCALP, IALP, etc.). Thus, similar to sTNALP discussed above, the present disclosure also provides other soluble human ALP isozymes, i.e., without the peptide signal, preferably comprising the extracellular domain of the ALPs. The sALPs of the invention also include polypeptide sequences satisfying a consensus sequence derived from the ALP extracellular domain of human ALP isozymes and of mammalian TNALP orthologs (human, mouse, rat, cow, cat, and dog) or a consensus derived from the ALP extracellular domain of just mammalian TNALP orthologs (human, mouse, rat, cow, cat, and dog). The sALPs of the invention also include those which satisfy similar consensus sequences derived from various combinations of these TNALP orthologs or human ALP isozymes. Such consensus sequences are given, for example, in WO 2008/138131.

sALPs of the present invention can include not only the wild-type sequence of the sALPs described above, but any polypeptide having at least 50% (e.g., 55%, 60%, 65%, 70%, 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more) sequence identity to these alkaline phosphatases (e.g., SEQ ID NOs: 1-24; for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa). Examples of mutations that can be introduced into an ALP sequence are described in US Publication No. 2013/0323244, hereby incorporated by reference in its entirety. A sALP can optionally be glycosylated at any appropriate one or more amino acid residues. In addition, an sALP can have at least 50% (e.g., 55%, 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more) sequence identity to any of the sALPs described herein (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa). A sALP can have 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more additions, deletions, or substitutions relative to any of the sALPs described herein (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa).

sALP Fusion Polypeptides

Any of the sALPs and linkers described herein can be combined in a sALP polypeptide, e.g., a sALP polypeptide of A-sALP-B, wherein each of A and B is absent or is an amino acid sequence of at least one amino acid (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa). When present, A and/or B can be any linker described herein. In some sALP polypeptides, A is absent, B is absent, or A and B are both absent. The sALP polypeptides of the invention can optionally include an Fc region to provide an sALP fusion polypeptide, as described herein. The sALP polypeptide can optionally include a bone-targeting moiety, as described herein. In some sALP polypeptides, a linker, e.g., a flexible linker, can be included between the bone-targeting moiety and the sALP, such as a dipeptide sequence (e.g., leucine-lysine or aspartic acid-isoleucine). Further exemplary Fc regions, linkers, and bone-targeting moieties are described below.

Any of the sALPs, linkers, and Fc regions described herein can be combined in a fusion polypeptide, e.g., a recombinant fusion polypeptide, which includes the structure Z-sALP-Y-spacer-X-Wn-V, Z-Wn-X-spacer-Y-sALP-V, Z-sALP-Y-Wn-X-spacer-V, and Z-Wn-X-sALP-Y-spacer-V (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa). In particular, the structure can be Z-sALP-Y-spacer-X-Wn-V or Z-Wn-X-spacer-Y-sALP-V. The sALP can be the full-length or functional fragments of ALPs, such as the soluble, extracellular domain of the ALP, as is described herein (e.g., TNALP, PALP, GCALP and IALP). Any one of X, Y, Z, and V and/or the spacer can be absent or an amino acid sequence of at least one amino acid. Wn can be a bone-targeting moiety, e.g., having a series of consecutive Asp or Glu residues, in which n=1 to 50, e.g., n=3-30, e.g., 5-15, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 36, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50. The bone-targeting moiety, if present, can be positioned anywhere in the fusion polypeptide, e.g., at or near the N-terminal or C-terminal end, and/or in the linker region. For instance, the bone-targeting moiety is at the C-terminal end. sALP polypeptides and fusion polypeptides can also not include a bone-targeting moiety.

sALP fusion polypeptides of the present invention can be of the structure hTNALP-Fc-D10. In particular, sALP fusion polypeptides can include an amino acid sequence of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa.

Useful spacers include, but are not limited to, polypeptides comprising a Fc, and hydrophilic and flexible polypeptides able to alleviate the repulsive forces caused by the presence of the terminal highly negatively charged peptide (e.g., Wn). For example, a sALP can be a fusion polypeptide including an Fc region of an immunoglobulin at the N-terminal or C-terminal domain. An immunoglobulin molecule has a structure that is well known in the art. It includes two light chains (˜23 kD each) and two heavy chains (˜50-70 kD each) joined by inter-chain disulfide bonds. Immunoglobulins are readily cleaved proteolytically (e.g., by papain cleavage) into Fab (containing the light chain and the VH and CH1 domains of the heavy chain) and Fc (containing the CH2 and CH3 domains of the heavy chain, along with adjoining sequences). Useful Fc fragments as described herein include the Fc fragment of any immunoglobulin molecule, including IgG, IgM, IgA, IgD, or IgE, and their various subclasses (e.g., IgG-1, IgG-2, IgG-3, IgG-4, IgA-1, IgA-2), from any mammal (e.g., human). For instance, the Fc fragment is human IgG-1. The Fc fragments of the invention can include, for example, the CH2 and CH3 domains of the heavy chain and any portion of the hinge region. The Fc region can optionally be glycosylated at any appropriate one or more amino acid residues known to those skilled in the art. In particular, the Fc fragment of the fusion polypeptide has the amino acid sequence of SEQ ID NO: 20, or has at least 50% (e.g., 55%, 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more) sequence identity to SEQ ID NO: 20. Engineered, e.g., non-naturally occurring, Fc regions can be utilized in the methods of the invention, e.g., as described in International Application Pub. No. WO2005/007809, which is hereby incorporated by reference. An Fc fragment as described herein can have 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 50, or more additions, deletions, or substitutions relative to any of the Fc fragments described herein.

The sALP fusion polypeptides described herein (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can include a peptide linker region between the Fc fragment. In addition, a peptide linker region can be included between the Fc fragment and the optional bone-targeting moiety. The linker region can be of any sequence and length that allows the sALP to remain biologically active, e.g., not sterically hindered. Exemplary linker lengths are between 1 and 200 amino acid residues, e.g., 1-5, 6-10, 11-15, 16-20, 21-25, 26-30, 31-35, 36-40, 41-45, 46-50, 51-55, 56-60, 61-65, 66-70, 71-75, 76-80, 81-85, 86-90, 91-95, 96-100, 101-110, 111-120, 121-130, 131-140, 141-150, 151-160, 161-170, 171-180, 181-190, or 191-200 amino acid residues. For instance, linkers include or consist of flexible portions, e.g., regions without significant fixed secondary or tertiary structure. Exemplary flexible linkers are glycine-rich linkers, e.g., containing at least 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or even 100% glycine residues. Linkers can also contain, e.g., serine residues. In some cases, the amino acid sequence of linkers consists only of glycine and serine residues. A linker can optionally be glycosylated at any appropriate one or more amino acid residues. Additionally, a linker as described herein can include any other sequence or moiety, attached covalently or non-covalently. The linker can also be absent, in which the Fc fragment and the sALP are fused together directly, with no intervening residues. Certain Fc-sALP or sALP-Fc fusion polypeptides can be viewed, according to the present disclosure, either as 1) having no linker, or as 2) having a linker which corresponds to a portion of the sALP. For example, Fc fused directly to hsTNALP (1-502) can be viewed, e.g., either as having no linker, in which the hsTNALP is amino acids 1-502, or as having a 17-amino acid linker, in which the hsTNALP (18-502).

Additional amino acid residues can be introduced into the polypeptide according to the cloning strategy used to produce the fusion polypeptides. For instance, the additional amino acid residues do not provide an additional GPI anchoring signal so as to maintain the polypeptide in a soluble form. Furthermore, any such additional amino acid residues, when incorporated into the polypeptide of the invention, do not provide a cleavage site for endoproteases of the host cell. The likelihood that a designed sequence would be cleaved by the endoproteases of the host cell can be predicted as described, e.g., by Ikezawa (Biol. Pharm. Bull. 25:409-417, 2002).

The sALPs and sALP fusion polypeptides of the invention (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can be associated into dimers or tetramers. For example, two sALP-Fc monomers can covalently be linked through two disulfide bonds located in the hinge regions of the Fc fragments. Additionally, the polypeptide or fusion polypeptide of the invention (e.g., a sALP polypeptide or fusion polypeptide) can be glycosylated or PEGylated.

Production of Nucleic Acids and Polypeptides

The nucleic acids encoding sALPs and sALP fusion polypeptides of the invention (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can be produced by any method known in the art. Typically, a nucleic acid encoding the desired fusion polypeptide is generated using molecular cloning methods, and is generally placed within a vector, such as a plasmid or virus. The vector is used to transform the nucleic acid into a host cell appropriate for the expression of the fusion polypeptide. Representative methods are disclosed, for example, in Maniatis et al. (Cold Springs Harbor Laboratory, 1989). Many cell types can be used as appropriate host cells, although mammalian cells are preferable because they are able to confer appropriate post-translational modifications. Host cells of the present invention can include, e.g., Chinese Hamster Ovary (CHO) cell, L cell, C127 cell, 3T3 cell, BHK cell, COS-7 cell or any other suitable host cell known in the art. For example, the host cell is a Chinese Hamster Ovary (CHO) cell (e.g., a CHO-DG44 cell).

The sALPs and sALP fusion polypeptides (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can be produced under any conditions suitable to effect expression of the sALP polypeptide in the host cell. Such conditions include appropriate selection of a media prepared with components such as a buffer, bicarbonate and/or HEPES, ions like chloride, phosphate, calcium, sodium, potassium, magnesium, iron, carbon sources like simple sugars, amino acids, potentially lipids, nucleotides, vitamins and growth factors like insulin; regular commercially available media like alpha-MEM, DMEM, Ham's-F12, and IMDM supplemented with 2-4 mM L-glutamine and 5% Fetal bovine serum; regular commercially available animal protein free media like Hyclone™ SFM4CHO, Sigma CHO DHFR, Cambrex POW ER™ CHO CD supplemented with 2-4 mM L-glutamine. These media are desirably prepared without thymidine, hypoxanthine and L-glycine to maintain selective pressure, allowing stable protein-product expression.

Pharmaceutical Compositions and Formulations

A composition of the present invention (e.g., including a sALP or sALP fusion polypeptide, such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can be administered by a variety of methods known in the art. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results. The route of administration can depend on a variety of factors, such as the environment and therapeutic goals. In particular, the polypeptides and fusion polypeptides described herein can be administration by any route known in the art, e.g., subcutaneous (e.g., by subcutaneous injection), intravenously, orally, nasally, intramuscularly, sublingually, intrathecally, or intradermally, or by combinations thereof. By way of example, pharmaceutical compositions of the invention can be in the form of a liquid, solution, suspension, pill, capsule, tablet, gelcap, powder, gel, ointment, cream, nebulae, mist, atomized vapor, aerosol, or phytosome.

Dosage

Any amount of a pharmaceutical composition (e.g., including a sALP or sALP fusion polypeptide, such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can be administered to a child having HPP (e.g., a child of about 5 to about 12 years of age). The dosages will depend on many factors including the mode of administration and the age of the patient. Typically, the amount of the composition (e.g., a sALP or sALP fusion polypeptide, such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) contained within a single dose will be an amount that is effective to treat a condition (e.g., HPP) as described herein without inducing significant toxicity.

For example, the sALP polypeptides (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) described herein can be administered to an HPP patient, such as a child having HPP (e.g., a child of about 5 to about 12 years of age) or an infant having HPP (e.g., an infant of about 3 years of age or less than 3 years of age), in individual doses ranging, e.g., from 0.01 mg/kg to 500 mg/kg (e.g., from 0.05 mg/kg to 500 mg/kg, from 0.1 mg/kg to 20 mg/kg, from 5 mg/kg to 500 mg/kg, from 0.1 mg/kg to 100 mg/kg, from 10 mg/kg to 100 mg/kg, from 0.1 mg/kg to 50 mg/kg, 0.5 mg/kg to 25 mg/kg, 1.0 mg/kg to 10 mg/kg, 1.5 mg/kg to 5 mg/kg, or 2.0 mg/kg to 3.0 mg/kg) or from 1 μg/kg to 1,000 μg/kg (e.g., from 5 μg/kg to 1,000 μg/kg, from 1 μg/kg to 750 μg/kg, from 5 μg/kg to 750 μg/kg, from 10 μg/kg to 750 μg/kg, from 1 μg/kg to 500 μg/kg, from 5 μg/kg to 500 μg/kg, from 10 μg/kg to 500 μg/kg, from 1 μg/kg to 100 μg/kg, from 5 μg/kg to 100 μg/kg, from 10 μg/kg to 100 μg/kg, from 1 μg/kg to 50 μg/kg, from 5 μg/kg to 50 μg/kg, or from 10 μg/kg to 50 μg/kg).

Exemplary doses of a sALP include, e.g., 0.01, 0.05, 0.1, 0.5, 1, 2, 2.5, 5, 10, 20, 25, 50, 100, 125, 150, 200, 250, or 500 mg/kg; or 1, 2, 2.5, 5, 10, 20, 25, 50, 100, 125, 150, 200, 250, 500, 750, 900, or 1,000 μg/kg. For all dosages or ranges recited herein, the term “about” can be used to modify these dosages by ±10% of the recited values or range endpoints. In particular, compositions (e.g., including sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa)) in accordance with the present disclosure can be administered to patients in doses ranging from about 0.001 mg/kg/day to about 500 mg/kg/day, about 0.01 mg/kg/day to about 100 mg/kg/day, or about 0.01 mg/kg/day to about 20 mg/kg/day. For example, the sALP compositions (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can be administered to patients in a weekly dosage ranging, e.g., from about 0.5 mg/kg/week to about 140 mg/kg/week, e.g., about 0.8 mg/kg/week to about 50 mg/kg/week, or about 1 mg/kg/week to about 10 mg/kg/week (e.g., about 6 or about 9 mg/kg/week). In particular, the sALP (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can be administered at a dosage of 2 mg/kg three times a week (total dose 6 mg/kg/week), 1 mg/kg six times a week (total dose 6 mg/kg/week), 3 mg/kg three times a week (total dose 9 mg/kg/week), 0.5 mg/kg three times a week (total dose of 1.5 mg/kg/week), or 9.3 mg/kg three times a week (total dose 28 mg/kg/week). The dosage will be adapted by the clinician in accordance with conventional factors such as the extent of the disease and different parameters from the HPP patient, such as a child having HPP (e.g., a child of about 5 to about 12 years of age) or an infant having HPP (e.g., an infant of about 3 years of age or less than 3 years of age).

Dosages of compositions including sALPs and sALP fusion polypeptides (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can be provided in either a single or multiple dosage regimens. Doses can be administered, e.g., hourly, bihourly, daily, bidaily, twice a week, three times a week, four times a week, five times a week, six times a week, weekly, biweekly, monthly, bimonthly, or yearly. Alternatively, doses can be administered, e.g., twice, three times, four times, five times, six times, seven times, eight times, nine times, 10 times, 11 times, or 12 times per day. In particular, the dosing regimen is once weekly. The duration of the dosing regimen can be, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 day(s), week(s), or month(s), or even for the remaining lifespan of the HPP patient, such as a child having HPP (e.g., a child of about 5 to about 12 years of age) or an infant having HPP (e.g., an infant of about 3 years of age or less than 3 years of age). The amount, frequency, and duration of dosage will be adapted by the clinician in accordance with conventional factors such as the extent of the disease and different parameters from the HPP patient, such as a child having HPP (e.g., a child of about 5 to about 12 years of age) or an infant having HPP (e.g., an infant of about 3 years of age or less than 3 years of age).

For example, a sALP or sALP fusion polypeptide (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can be formulated as a solution for injection, which is a clear, colorless to slightly yellow, aqueous solution, pH 7.4. The sALP or sALP polypeptide (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) may be formulated at a concentration of 12 mg/0.3 mL, 18 mg/0.45 mL, 28 mg/0.7 mL, 40 mg/1 ml, or 80 mg/0.8 mL. In particular, the composition can be formulated as a 40 mg/ml solution for injection, in which each ml of solution contains 40 mg of sALP or sALP polypeptide (e.g., each vial contains 0.3 ml solution and 12 mg of sALP (40 mg/ml), each vial contains 0.45 ml solution and 18 mg of sALP (40 mg/ml), each vial contains 0.7 ml solution and 28 mg of sALP (40 mg/ml), or each vial contains 1.0 ml solution and 40 mg of asfotase alfa (40 mg/ml)). A sALP or sALP polypeptide (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can be formulated as a solution for injection at a concentration of 100 mg/ml, in which each 1 ml of solution contains 100 mg of sALP or sALP polypeptide (e.g., each vial contains 0.8 ml solution and 80 mg of asfotase alfa (100 mg/ml)).

For example, the recommended dosage of a sALP or sALP fusion polypeptide ((such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) is 2 mg/kg of body weight administered subcutaneously three times per week, or a dosage regimen of 1 mg/kg of body weight administered subcutaneously six times per week. Additional dosage information is provided below (Table 1).

TABLE 1 Dosing of asfotase alfa. If injecting 3x per week If injecting 6 x per week Body Dose Volume Vial type Dose to Volume Vial type Weight to be to be used for be to be used for (kg) injected injected injection injected injected injection 3 6 mg 0.15 ml 0.3 ml 4 8 mg 0.20 ml 0.3 ml 5 10 mg 0.25 ml 0.3 ml 6 12 mg 0.30 ml 0.3 ml 6 mg 0.15 ml 0.3 ml 7 14 mg 0.35 ml 0.45 ml 7 mg 0.18 ml 0.3 ml 8 16 mg 0.40 ml 0.45 ml 8 mg 0.20 ml 0.3 ml 9 18 mg 0.45 ml 0.45 ml 9 mg 0.23 ml 0.3 ml 10 20 mg 0.50 ml 0.7 ml 10 mg 0.25 ml 0.3 ml 11 22 mg 0.55 ml 0.7 ml 11 mg 0.28 ml 0.3 ml 12 24 mg 0.60 ml 0.7 ml 12 mg 0.30 ml 0.3 ml 13 26 mg 0.65 ml 0.7 ml 13 mg 0.33 ml 0.45 ml 14 28 mg 0.70 ml 0.7 ml 14 mg 0.35 ml 0.45 ml 15 30 mg 0.75 ml 1 ml 15 mg 0.38 ml 0.45 ml 16 32 mg 0.80 ml 1 ml 16 mg 0.40 ml 0.45 ml 17 34 mg 0.85 ml 1 ml 17 mg 0.43 ml 0.45 ml 18 36 mg 0.90 ml 1 ml 18 mg 0.45 ml 0.45 ml 19 38 mg 0.95 ml 1 ml 19 mg 0.48 ml 0.7 ml 20 40 mg 1.00 ml 1 ml 20 mg 0.50 ml 0.7 ml 25 50 mg 0.50 ml 0.8 ml 25 mg 0.63 ml 0.7 ml 30 60 mg 0.60 ml 0.8 ml 30 mg 0.75 ml 1 ml 35 70 mg 0.70 ml 0.8 ml 35 mg 0.88 ml 1 ml 40 80 mg 0.80 ml 0.8 ml 40 mg 1.00 ml 1 ml 50 50 mg 0.50 ml 0.8 ml 60 60 mg 0.60 ml 0.8 ml 70 70 mg 0.70 ml 0.8 ml 80 80 mg 0.80 ml 0.8 ml 90 90 mg 0.90 ml 0.8 ml (x2) 100 100 mg 1.00 ml 0.8 ml (x2)

Formulations

The compositions including sALPs and sALP fusion polypeptides (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can be formulated according to standard methods. Pharmaceutical formulation is a well-established art, and is further described in, e.g., Gennaro (2000) “Remington: The Science and Practice of Pharmacy,” 20th Edition, Lippincott, Williams & Wilkins (ISBN: 0683306472); Ansel et al. (1999) “Pharmaceutical Dosage Forms and Drug Delivery Systems,” 7th Edition, Lippincott Williams & Wilkins Publishers (ISBN: 0683305727); and Kibbe (2000) “Handbook of Pharmaceutical Excipients American Pharmaceutical Association,” 3rd Edition (ISBN: 091733096X). For instance, a sALP composition (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can be formulated, for example, as a buffered solution at a suitable concentration and suitable for storage at 2-8° C. (e.g., 4° C.). A composition can also be formulated for storage at a temperature below 0° C. (e.g., −20° C. or −80° C.). A composition can further be formulated for storage for up to 2 years (e.g., one month, two months, three months, four months, five months, six months, seven months, eight months, nine months, 10 months, 11 months, 1 year, 1½ years, or 2 years) at 2-8° C. (e.g., 4° C.). Thus, the compositions described herein can be stable in storage for at least 1 year at 2-8° C. (e.g., 4° C.).

The compositions including sALPs and sALP fusion polypeptides (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can be in a variety of forms. These forms include, e.g., liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, tablets, pills, powders, liposomes and suppositories. The preferred form depends, in part, on the intended mode of administration and therapeutic application.

For example, compositions intended for systemic or local delivery can be in the form of injectable or infusible solutions. Accordingly, the compositions (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can be formulated for administration by a parenteral mode (e.g., subcutaneous, intravenous, intraperitoneal, or intramuscular injection). “Parenteral administration,” “administered parenterally,” and other grammatically equivalent phrases, as used herein, refer to modes of administration other than enteral and topical administration, usually by injection, and include, without limitation, subcutaneous, intradermal, intravenous, intranasal, intraocular, pulmonary, intramuscular, intra-arterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intrapulmonary, intraperitoneal, transtracheal, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural, intracerebral, intracranial, intracarotid, and intrasternal injection and infusion.

The compositions including sALPs and sALP fusion polypeptides (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can be formulated as a solution, microemulsion, dispersion, liposome, or other ordered structure suitable for stable storage at high concentration. Sterile injectable solutions can be prepared by incorporating a composition described herein in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filter sterilization. Generally, dispersions are prepared by incorporating a composition described herein into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, methods for preparation include vacuum drying and freeze-drying that yield a powder of a composition described herein plus any additional desired ingredient (see below) from a previously sterile-filtered solution thereof. The proper fluidity of a solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prolonged absorption of injectable compositions can be brought about by including in the composition a reagent that delays absorption, for example, monostearate salts, and gelatin.

The compositions described herein can also be formulated in immunoliposome compositions. Such formulations can be prepared by methods known in the art such as, e.g., the methods described in Epstein et al. (1985) Proc Natl Acad Sci USA 82:3688; Hwang et al. (1980) Proc Natl Acad Sci USA 77:4030; and U.S. Pat. Nos. 4,485,045 and 4,544,545. Liposomes with enhanced circulation time are disclosed in, e.g., U.S. Pat. No. 5,013,556.

Compositions including sALPs and sALP fusion polypeptides (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can also be formulated with a carrier that will protect the composition (e.g., a sALP polypeptide or sALP fusion polypeptide) against rapid release, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are known in the art. See, e.g., J. R. Robinson (1978) “Sustained and Controlled Release Drug Delivery Systems,” Marcel Dekker, Inc., New York.

When compositions are to be used in combination with a second active agent, the compositions can be co-formulated with the second agent, or the compositions can be formulated separately from the second agent formulation. For example, the respective pharmaceutical compositions can be mixed, e.g., just prior to administration, and administered together or can be administered separately, e.g., at the same or different times.

Compositions including sALPs and sALP fusion polypeptides (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can be formulated for administration to a patient or, if administered to a fetus, to a female carrying such fetus, along with intravenous gamma globulin therapy (IVIG), plasmapheresis, plasma replacement, or plasma exchange.

Carriers/Vehicles

Preparations containing a sALP or sALP fusion polypeptide (such as TNALP, for example the sALP polypeptide of SEQ ID NO: 1 or a polypeptide variant having at least 95% sequence identity to the sequence of SEQ ID NO: 1, e.g., asfotase alfa) can be provided to HPP patients, such as a child having HPP (e.g., a child of about 5 to about 12 years of age) or an infant having HPP (e.g., an infant of about 3 years of age or less than 3 years of age), in combination with pharmaceutically acceptable sterile aqueous or non-aqueous solvents, suspensions or emulsions. Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oil, fish oil, and injectable organic esters. Aqueous carriers include water, water-alcohol solutions, emulsions or suspensions, including saline and buffered medical parenteral vehicles including sodium chloride solution, Ringer's dextrose solution, dextrose plus sodium chloride solution, Ringer's solution containing lactose, or fixed oils. For example, the pharmaceutically acceptable carrier can include sodium chloride and/or sodium phosphate, in which the composition includes, e.g., about 150 mM sodium chloride and/or about 25 mM sodium phosphate, pH 7.4.

Intravenous vehicles can include fluid and nutrient replenishers, electrolyte replenishers, such as those based upon Ringer's dextrose, and the like. Pharmaceutically acceptable salts can be included therein, for example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates, and the like. Additionally, auxiliary substances, such as wetting or emulsifying agents, pH buffering substances, and the like, can be present in such vehicles. A thorough discussion of pharmaceutically acceptable carriers is available in Remington's Pharmaceutical Sciences (Mack Pub. Co., N.J. 1991).

The following examples are intended to illustrate, rather than limit, the disclosure. These studies feature the administration of asfotase alfa (SEQ ID NO: 1) to children of about 5 to about 12 years of age to treat HPP, its symptoms, and physical impairments associated therewith for an extended period of time.

Example 1. Study Design for Treatment of HPP Children with Asfotase Alfa

Children with hypophosphatasia (HPP) of about 5 years to about 12 years of age participated in an initial phase study to determine the efficacy, safety, tolerability, and pharmacokinetics of treatment with a soluble alkaline phosphatase (sALP; asfotase alfa; SEQ ID NO: 1) for 6 months (Table 2). Inclusion criteria included radiographic skeletal features of hypophosphatasia-related rickets, serum ALP activity below age-adjusted lower limits of normal, and plasma PLP levels at least twice the upper limit of normal. Serum 25-hydroxyvitamin D had to be greater than or equal to 20 ng/ml (50 nM). Exclusion criteria included treatable rickets, hypocalcemia, hypophosphatemia, or bisphosphonate exposure.

TABLE 2 Baseline characteristics of historical control HPP patients and HPP patients treated with asfotase alfa. Historical Control HPP Patients Treated Patients n 16 13 Age (yrs) at enrollment Mean ± SD 6.0 ± 1.8 8.8 ± 2.2 Median (min, max) 5.5 (4, 11) 8.6 (6, 12) Sex, % (n) Male 69% (11) 85% (11) Ethnicity, % (n) Not Hispanic or Latino NA 92% (12) Race, % (n) White NA 92% (12) Form of hypophosphatasia, % (n) Infantile 44% (7) 38% (5) Childhood (≥6 mos to <18 yrs) 56% (9) 62% (8) Age (mos) at onset of hypophosphatasia symptoms Mean ± SD 7.4 ± 9.5 10.5 ± 7.0  Median (min, max) 6.0 (0, 40) 12.0 (1, 22) Baseline RSS (0 = normal, 10 = severe) Mean ± SD 1.44 ± 0.96 2.77 ± 1.33 Median (min, max) 1.0 (0.0, 3.5) 3.0 (0.5, 6.0) Baseline plasma PPi (μM) Mean ± SD NA 5.01 ± 0.97 Median (min, max [normal range]) 4.86 (3.74, 6.96 [0.75-5.71 μM]) Baseline serum PLP (ng/ml) Mean ± SD 323 ± 178 214 ± 127 Median (min, max [normal range]) 328 (85, 726 [5.7-61.2 218 (76, 527 [5.7-61.2 ng/ml]) ng/ml]) Baseline serum calciump (mmol/l) Mean ± SD ± ± Median (min, max [normal range]) 2.52 (2.35, 2.78 [2.12-2.57 2.50 (2.37, 2.67 [2.12-2.57 mmol/l]) mmol/l]) Hypophosphatasia-related disease history, % (n) Unusual gait or running NA 100% (13) Premature tooth loss 100% (13) Delayed (≥15 mos) walking 85% (11) Knock knees 77% (10) Muscle weakness 62% (8) Elevated serum phosphorous 54% (7) Difficulty eating/swallowing 46% (6) Difficulty gaining weight 46% (6) Hypermobility 46% (6) Joint pain 46% (6) Muscle pain 46% (6) Abnormally shaped chest 46% (6) Bone pain severe enough to limit activities 46% (6) Bone pain severe enough to require medication 39% (5) Bowing of legs 39% (5)

A total of 13 HPP patients treated with asfotase alfa (40 or 100 mg/ml concentration) were initially randomized to receive 2 mg/kg or 3 mg/kg three times weekly (i.e., 6 or 9 mg/kg/week) for 6 months (FIG. 1). In the extension study of at least 5 years, HPP patients initially received asfotase alfa administered via subcutaneous administration 3 mg/kg/week, either as 0.5 mg/kg six times weekly or 1 mg/kg three times weekly, until preliminary analysis led to a protocol amendment that doubled the dose to 6 mg/kg/week after three to nine months to maintain therapeutic efficacy. Since a separately identified historical control patient group was used for comparison, there was no randomization between the patients treated with asfotase alfa and historical control HPP patients.

Skeletal abnormalities of HPP children at baseline included knock knees (77%), rachitic chest (46%), bowing of legs (39%), and craniosynostosis (31%) and were accompanied by muscle weakness in most patients and an unusual gait in all patients. Radiographic abnormalities observed in children with HPP at baseline included metaphyseal flaring, metadiaphyseal sclerosis, radiolucencies, apparent physical widening, irregularity of the provisional zone of calcification, and transverse subphyseal band of lucency (FIG. 2). Bone pain often limited activities (46%) and/or required analgesics (39%). Nearly half of the patients had poor weight gain and prior difficulty in feeding. Hypercalcemia occurred in 31% of patients. After 1 month of treatment, 1 boy withdrew for elective scoliosis surgery.

Patients treated with asfotase alfa were assessed for skeletal manifestations of HPP using the Radiographic Global Impression of Change (RGI-C) and Rickets Severity Score (RSS); changes in tissue-nonspecific alkaline phosphatase (TNALP) substrate levels (plasma PPi and PLP); growth (height, weight, and body mass index (BMI) Z-scores); strength of muscle groups that move the hips and knees, measured using a hand-held dynamometer; walking ability and endurance or speed using a Six Minute Walk Test (6MWT); physical function and strength using the Bruininks-Oseretsky Test of Motor Proficiency, 2nd Edition (BOT-2); global function, including pain and interference with normal activities, using the Pediatric Outcomes Data Collection Instrument (PODCI); and disability, using the Childhood Health Assessment Questionnaire (CHAQ).

An overview of the metrics used in this study to assess treatment of childhood HPP with asfotase alfa over an extended period (≥5 years of treatment) is shown in Table 3. In particular, physical function and impairments of HPP patients were assessed with the Bruininks-Oseretsky Test of Motor Proficiency, 2nd Edition (BOT-2). The BOT-2 was administered to capture body structure impairments and mobility restrictions important in HPP in the BOT-2 subtests of strength and running speed and agility, as compared to a normal age-matched population with a standard BOT-2 score of 15 (standard deviation of 5).

Ability to perform Activities of Daily Living (ADL) and pain of HPP patients were assessed with the Childhood Health Assessment Questionnaire (CHAQ) and the Pediatric Outcomes Data Collection Instrument (PODCI). The CHAQ has two scales: a Disability Index with 30 age-appropriate items in 8 subscales of ADL, and a Discomfort (pain) Index. CHAQ disability is scored from 0 to 3 (0=functional independence; 3=complete dependence on caregiver). For example, a decrease of ≥0.13 in a CHAQ index score is considered clinically meaningful in juvenile arthritis (see Dempster et al., Arthritis Rheum. 44(8): 1768-74, 2001; incorporated herein by reference). The PODCI has eight scales, including Upper Extremity and Physical Function, Transfer and Basic Mobility, Sports and Physical Functioning, and Global Functioning; each has a normative mean score for healthy patients of 50 (standard deviation of 10).

TABLE 3 Metrics used to evaluate physical function, activities of daily living, and pain of HPP patients. Physical Function Activities of Daily Living/Pain BOT-2 Strength and Agility Childhood Health Assessment Questionnaire (CHAQ) Assesses motor ability in Disability index, includes 8 sub-scales: 4-21 year olds Dressing/grooming Valid reliable, standardized Arising tool with normative data Eating Captures body structure Walking impairments and mobility Hygiene restrictions important in HPP Reach Strength Grip Running Speed and Activities Agility Score 0-3 where 0 = no difficulty and 3 = unable to Standard Score: do Mean 15, SD 5 Discomfort (Pain) Index, visual analog sale (single Scores compared with question) normal age-matched Pediatric Outcomes Data Collection Instrument (PODCI) population 8 scales: Transfer and basic mobility Sports and physical functioning Pain and comfort Upper extremity and physical function Treatment expectations Happiness Satisfaction with symptoms Global functioning scale Standardized scale from 0 to 100, 100 = less disability

Example 2. Skeletal Changes in HPP Children Treated with Asfotase Alfa

Radiographic assessments of HPP patients were performed during the course of treatment as previously described (see Whyte et al. (N Engl J Med 366(10): 904-913, 2012); incorporated herein by reference), with 16 historical controls. Radiographic skeletal changes during the first 2 years of asfotase alfa treatment were compared to the historical control HPP patients. Bilateral wrist and/or knee radiographs, previously obtained at intervals spanning 6 months to 2 years between 5 and 12 years of age, were evaluated as close to 6 months or 1 to 2 years apart for comparison with the treatment group.

The Radiographic Global Impression of Change (RGI-C), a 7-point ordinal scale, assesses HPP skeletal disease in wrists and knees, including physeal widening, irregularity of the provisional zone of calcification, metaphyseal flaring, radiolucencies, patchy osteosclerosis, and recent fractures. The changes compared with baseline were quantified independently by 3 pediatric radiologists blinded to treatment time points and group assignments. For the RGI-C, +3 signifies complete or nearly complete healing, whereas −3 represents severe worsening. The scores per patient were averaged across radiologists at each time point. An RGI-C “responder” had a score greater than or equal to +2 (i.e., substantial or near/complete healing).

The Rickets Severity Score (RSS), which was developed to assess nutritional rickets, was scored by the same individual rater to: (i) evaluate the radiographs of patients treated with asfotase alfa throughout the study and (ii) evaluate the historical control radiographs representing 6 month, 1 year, and 2 year intervals. Knee and wrist growth-plate abnormalities and metaphyseal fraying and cupping were rated according to a 10-point scale (0=no rickets; 10=severe rickets), with scores for each radiograph determined in random order (Table 4).

TABLE 4 Rickets Severity Score (RSS) 10-point radiographic scoring method for rickets. Wrist: score both radius and ulna separately Grade Radiographic features 0 Normal growth plate without changes of rickets 0.5 Lucency of metaphyseal margin without fraying or irregularity 1 Widened growth plate, irregularity of metaphyseal margin but without concave cupping 1.5 Partial metaphyseal concavity or incomplete fraying of metaphyseal margin 2 Metaphyseal concavity with fraying of margins 2 bones × 2 points = 4 points possible Knee: score both femur and tibia separately Multiply the grade in A by the multiplier in B for each bone, then add femur and tibia scores together A Grade Degree of lucency and widening of zone of provisional calcification 0 Normal growth plate without changes of rickets 1 Partial lucency, smooth margin of metaphysis visible 2 Partial lucency, smooth margin of metaphysis NOT visible 3 Complete lucency, epiphysis appears widely separated from distal met B Multiplier Portion of growth plate affected 0.5 ≤1 condyle or plateau 1 2 condyles or plateaus 2 bones × 1 point × 3 points = 6 points possible Total: 10 points possible that would indicate very severe rickets

The RGI-C median scores were compared using Wilcoxon rank-sum test. Missing values were imputed with last observation carried forward (LOCF), and patients with no post-baseline assessment were assigned “no change” (e.g., RGI-C=0). Observed median and mean within-group changes were also assessed for difference from 0 using the Wilcoxon signed-rank test and paired t-test. All analyses were 2-sided, with significance defined as a P-value less than 0.05.

After 6 weeks of treatment with asfotase alfa, the median RGI-C score had improved significantly (+1.0 [0.0, +2.0; min, max]; P=0.001). Further improvement occurred by 6 months and persisted through 5 years (+2.2 [+1.7, +2.7]; P=0.0005). In contrast, no significant change occurred in the RGI-C score of the historical controls spanning up to 2 years (FIG. 3).

The RGI-C responder analysis showed 69% ( 9/13) of asfotase alfa-treated HPP patients achieved “responder” designation (RGI-C score≥2) at 6 months of therapy (LOCF data imputation applied) compared with 6% ( 1/16) of the historical controls (P=0.007). The percentage of asfotase alfa-treated HPP patients considered responders then further improved steadily; 75% at 2 years, 88% at 3 years, and 92% at 5 years (1 patient had withdrawn at month 1). Notably, improvements at 6 months of treatment with asfotase alfa persisted after 5 years of treatment, as evidenced by representative radiographic changes in a male patient with HPP that was 6 years of age at baseline (FIG. 4). Consistent with the RGI-C findings, the RSS analysis confirmed improvement of rickets after treatment of the HPP children with asfotase alfa with significantly decreased median RSS scores comparing the first eligible radiographs with all subsequent evaluated time points in comparison to the historical controls (Table 5).

TABLE 5 Change in Rickets Severity Score (RSS) scores with asfotase alfa treatment. Variable Baseline Month 6 Year 1 Year 2 Year 3 Year 4 Year 5 Historical Control Patients N 16 16 16 15 RSS, mean (SD) 1.44 (0.96) 1.31 (1.05) 1.19 (1.03) 1.43 (1.19) RSS, median 1.00 1.25 1.00 1.00 (min, max) (0.0, 3.5) (0.0, 4.0) (0.0, 4.0) (0.5, 5.0) RSS Change from 0.00 −0.50 0.00 baseline, median (−1.0, 1.5) (−1.0, 1.5) (−1.5, 1.5) (min, max) PA Asfotase Alfa-treated Patients N 12 12 12 12 8 10 10 RSS, mean (SD) 2.75 (1.39) 1.04 (1.23) 1.25 (1.25) 1.04 (1.12) 0.56 (0.68) 0.40 (0.52) 0.15 (0.34) RSS, median 2.75 0.75 1.00 0.50 0.50 0.25 0.00 (min, max) (0.5, 6.0) (0.0, 4.5) (0.0, 4.5) (0.0, 4.0) (0.0, 2.0) (0.0, 1.5) (0.0, 1.0) RSS Change from −1.50 −1.25 −2.00 −2.50 −2.50 −2.75 baseline, median (−3.5, −0.5) (−3.0, 0.0) (−3.5, 0.5) (−4.0, 1.0) (−4.5, 0.0) (−5.0, −0.5) (min, max) PA 0.0005 0.001 0.002 NA NA NA PB 0.0008 0.007 0.0025 AP value based on within group Wilcoxon signed-rank test for change from baseline. BP value based on Willcoxon rank sum test comparing change from baseline of the treatment group with the historical control group. NA, not available.

Example 3. Tissue-Nonspecific Alkaline Phosphatase (TNALP) Activity in HPP Children Treated with Asfotase Alfa

Of the HPP children treated with asfotase alfa, 12 of the 13 treated children had at least 1 mutant TNSALP allele identified using PCR and DNA sequencing of all coding exons and splice sites (Table 6). One patient did not show a TNALP gene mutation despite extensive molecular investigations.

TABLE 6 TNALP gene mutation analysis by HPP patient treated with asfotase alfa. Patient AA Change 1 Nuc Change 1 Number Gene Mutation Class AA Change 2 Nuc Change 2 1 Compound ALA176THR C.526G > A heterozygote ASP294ALA C.881A > C 2 Compound ALA176THR C.526G > A heterozygote ASP294ALA C.881A > C 3 Heterozygous ASP378VAL C.1133A > T 4 Compound GLU191LYS C.571G > A heterozygote ASP337GLY C.1010A > G 5 Compound GLU191LYS C.571G > A heterozygote ASN417SER C.1250A > G 6 Compound GLU191LYS C.571G > A heterozygote ASP294ALA C.881A > C 7 Compound ALA176THR C.526G > A heterozygote ALA179THR C.535G > A 8 Compound GLU84ASP C.252G > C heterozygote VAL459MET C.1375G > A 9 Compound GLU191LYS C.571G > A heterozygote GLY334ASP C.1001G > A 10  Compound GLU191LYS C.571 G > A heterozygote ALA116THR C.346G > A 11A NA NA NA Compound GLU191LYS C.571G > A 12  heterozygote P.327DELPHE C.978 980DELCTT Compound ALA114THR C.340G > A 13  heterozygote GLY455SER C.1363G > A AA, amino acid; NA, not available; Nuc, nucleotide. ANo mutation was detected.

The median serum ALP activity of the treatment group at baseline was 49 IU/l (min, max: 27, 68) and then rapidly increased after treatment with asfotase alfa (FIG. 5A). After 5 years of therapy, serum ALP remained markedly elevated at 5747 IU/l (3,039, 9,959). Baseline plasma PPi median concentration, 4.9 μM (3.7, 7.0), was near the upper limit of the age-dependent reference range (0.75-5.71 μM). Baseline plasma PLP median concentration, 218 ng/ml (76, 527), was clearly elevated (normal 5.7-61.2 ng/ml) in the HPP children. At 6 weeks of treatment, the rapid and significant reduction of plasma PPi and PLP concentrations resulted in normal limits for most asfotase alfa-treated children (2.0 μM [1.3, 3.1] and 10.6 ng/ml [0.9, 40.3], respectively; FIG. 5B and FIG. 5C, respectively). Normal levels of PPi and PLP were sustained throughout the study for the HPP children treated with asfotase alfa.

Example 4. Growth and Strength of HPP Children Treated with Asfotase Alfa

After treatment with asfotase alfa, the first significant increase in the weight Z-scores of the HPP patients was identified at 6 weeks (baseline: −1.21 [−8.2, 2.3]; 6 weeks: −0.92 [−8.0, 2.1], P=0.0048); FIG. 6A). The first significant increase in the height Z-scores of the HPP patients treated with asfotase alfa was noted after 1.5 years (baseline: −1.26 [−6.6, 0.0]; 1.5 years: −0.87 [−6.6, −0.3], P=0.0257; FIG. 6B). The median BMI Z-score of the HPP patients treated with asfotase alfa had increased 5 years of treatment (baseline: −0.57 [−1.4, 2.4]; 5 years: 0.05 [−1.0, 2.2], P=0.0077; FIG. 6C). Most asfotase alfa-treated patients became stronger for their age and sex; e.g., right hip abduction baseline median at 13.6 lbs was 7.3 and 22.1 (min, max, respectively). Right hip abduction of the HPP patients treated with asfotase alfa was 50% of the right hip abduction predicted for healthy peers at baseline (FIG. 7A). At 6 months and 18.3 pounds, minimum and maximum values were 8.4 and 28.2, respectively (P=0.0018 or 60% predicted). At 5 years and 30.7 pounds, minimum and maximum values were 19.5 and 84.4, respectively (P=0.0079 or 91% predicted). Significant improvements were similarly documented for hip and knee extensors and knee flexors of HPP children treated with asfotase alfa (FIG. 7B-7D).

Example 5. Physical Function of HPP Children Treated with Asfotase Alfa Assessed Using the Six Minute Walk Test (6MWT)

Physical function and impairments of the HPP children treated with asfotase alfa were assessed using the Six Minute Walk Test (6MWT). Before treatment, gross motor delays and functional disability were documented in most HPP patients; e.g., 9 of the 13 had a subnormal 6MWT (<80% predicted for of age- and sex-matched peers). Patients improved by a walking distance of at least 20 meters (e.g., a minimal clinically important difference (MCID)) after treatment with asfotase alfa. After 5 years of treatment, 7 of 9 children achieved or surpassed 80% of the predicted distance (±2 SD mean), indicating normal ambulation.

6MWT values improved for individual HPP patients and the median 6MWT value of the HPP patients treated with asfotase alfa increased from about 350 meters at baseline to about 550 meters after 5 years of treatment with asfotase alfa (FIG. 8A). The percent predicted 6MWT distance median of the HPP patients treated with asfotase was within the normal range after 6 months of treatment, which was sustained for at least 5 years of treatment with asfotase alfa (FIG. 8B). The median 6MWT distance of the HPP patients increased from 61% of the predicted 6MWT distance for healthy children (age- and sex-matched) at baseline to 83% of the predicted median 6MWT distance after 5 years of treatment with asfotase alfa (FIG. 8B).

Clinical relevance of the 6MWT value was assessed via Pearson correlations between 6MWT and measures of skeletal disease using the RGI-C scale, the RSS, and ability to perform ADL as assessed by the CHAQ Disability Index and PODCI Global Function, Transfer and Basic Mobility, and Sports and Physical Functioning scaled scores. Correlation analysis of 127 data points revealed significant (p<0.001), moderate-to-strong linear relationships between distance walked (percent predicted for age, gender, and height) and RSS (r=0.73), CHAQ Disability Index (r=−0.57), and PODCI subscales, including Global Function (r=0.76), Transfer and Basic Mobility (r=0.69), and Sports and Physical Functioning (r=0.78). Changes in 6MWT and RGI-C scores showed weaker correlation (r=0.32; 68 data points; p<0.01).

Additionally, all patients requiring the use of assistive devices to walk prior to treatment with asfotase alfa exhibited an improvement after treatment with asfotase alfa, such as treatment with asfotase alfa for one year. In particular, patients progressed from a wheelchair to crutches, from a walker to a cane, from a wheeled walker to independent ambulation, and from a cane to independent ambulation.

These results indicate that the 6MWT is a valid, clinically relevant measure of disability and treatment outcomes for patients with HPP, particularly HPP patients treated with asfotase alfa. Importantly, asfotase alfa effectively restores physical function and walking ability in children with HPP, as shown by the increase in the 6MWT distance after treatment with asfotase alfa.

Example 6. Physical Function of HPP Children Treated with Asfotase Alfa Assessed Using the Bruininks-Oseretsky Test of Motor Proficiency, Second Edition (BOT-2)

Physical function and impairments of the HPP patients treated with asfotase alfa were assessed using the Bruininks-Oseretsky Test of Motor Proficiency, Second Edition (BOT-2) strength test and the BOT-2 running speed and agility test. BOT-2 tests to assess strength of the HPP patients included sit-ups, v-ups, standing long jump, wall sit, and push-ups (FIG. 9A). Strength scores were converted to strength scaled scores relative to a healthy age and gender matched reference population for each time interval (baseline, 6 months, 1 year, 2 year, 3 years, 4 years, and 5 years) (FIG. 9B). The strength scaled score mean for age-matched healthy patients was 15 with a standard deviation (SD) of 5, resulting in a range of 10 to 20 for the healthy reference population. At baseline, patients had physical impairments compared with healthy peers with a BOT-2 Strength score of 4 (minimum score of 1, maximum score of 13). BOT-2 strength scores significantly improved by 6 months of treatment, resulting in a BOT-2 score within the range of healthy patients, which was sustained throughout at least 5 years of treatment with asfotase alfa.

BOT-2 tests to assess running speed and agility of the HPP patients included the 50 foot shuttle run, sideways steps over balance beam, and one and two legged side hops (FIG. 10A). Running speed and agility scores were converted to running speed and agility scaled scores relative to all HPP patients in the study and the median score was determined at each time interval (baseline, 6 months, 1 year, 2 year, 3 years, 4 years, and 5 years). The scaled score mean for age-matched healthy patients was 15 with a standard deviation (SD) of 5, resulting in a range of 10 to 20 for healthy patients. At baseline, patients had physical impairments compared to healthy peers with a BOT-2 running speed and agility score of 3 (minimum score of 1, maximum score of 9). BOT-2 running speed and agility scores significantly improved by 6 months of treatment, resulting in a BOT-2 score within the range of healthy patients by 3 years, which was sustained throughout 5 years of treatment with asfotase alfa (FIG. 10B).

Composite strength and agility scaled scores were also determined from the BOT-2 strength and running speed and agility for the HPP children treated with asfotase alfa relative to healthy children (FIG. 10C). The median value for the BOT-2 strength and agility composite score at baseline (28.0 [20.0, 37.0]; percentile rank=1.0) was greater than 2 SD below the median score of 50 for healthy, age-matched peers. During treatment, the BOT-2 strength and agility composite score improved, reaching the normal range after 1 year, and was significantly better after 5 years (46.0 [33.0, 64.0]; P≤0.0001; percentile rank=35.0; FIG. 10D). The magnitude of change in the standard scores indicated gross motor function within the normal range (±1 SD mean).

Example 7. Ability to Perform Activities of Daily Living and Pain of HPP Children Treated with Asfotase Alfa

The ability to perform activities of daily living (ADL) and pain of the HPP patients were reported by parents via the Childhood Health Assessment Questionnaire (CHAQ; Table 7). The CHAQ has a Disability Index with 30 age-appropriate items in 8 subscales (0-3, 0=no disability for the index) and a Discomfort (pain) Index (0-100, 0=no pain for the index).

TABLE 7 Description, scoring, and interpretation of CHAQ Disability and Discomfort tests. Component Disability Discomfort Description Covers major aspects of daily living Measures discomfort as determined by the (30 questions in 8 domains): presence of pain Dressing and grooming Visual analog scale (single question) Arising Eating Walking Hygiene Reach Grip Activities Scoring 0 = no difficulty 0-100 (anchored by 0, no pain; 100, very severe 1 = some difficulty pain) 2 = much difficulty 3 = unable to do Interpretation Decreased score = reduced disability Decreased score = reduced pain MCID: decrease ≥0.13 MCID not determined Healthy children (no disability) score 0

Ability to perform ADL and pain of the HPP children treated with asfotase alfa were also reported by parents using the Pediatric Outcomes Data Collection Instrument (PODCI). The PODCI has 8 scales, including Transfer and Basic Mobility, Sports and Physical Function, and Pain and Comfort (Table 8). For the PODCI, the normative mean was 50 (standard deviation of 10).

TABLE 8 Description of measurements and exemplary tasks of PODCI Transfer and Basic Mobility, Sports and Physical Function, and Pain and Comfort scales. Domain Example tasks Transfer and Basic Measures difficulty Climb one flight of stairs Mobility Scale experienced in performing Walk one block routine motion and motor Get on and off a bus activities in daily activities Bend over from a standing position and pick up something off the floor Sports/Physical Measures difficulty or Run short distances Functioning Scale limitations encountered in Climb three flights of stairs participating in more active How often in the last week did your activities or sports child participate in gym/recess? Pain/Comfort Scale Measures the level of pain Did pain or discomfort interfere with experienced during the past your child's activities? week During the last week, how much did pain interfere with your child's normal activities (including at home, outside of home, and at school)?

Pain experienced by the HPP patients decreased after administration of asfotase alfa with significant improvements observed by 2 years with an average CHAQ median disability index score of 0 (FIG. 11). This decrease in physical disability was sustained throughout 5 years with a CHAQ median disability index score of about 0. In particular, the elevated CHAQ pain (20.0 [0.0, 72.0]) and disability (1.0 [0.0, 2.3]) medians at baseline were 0 at 5 years of treatment with asfotase alfa, indicating no pain or disability for most of the HPP children (0.0 [0.0, 60.0], P=0.1125; 0.0 [0.0, 1.0], P=0.0002, respectively).

Likewise, physical disability of the HPP patients decreased after administration of asfotase alfa with significant improvements observed by 6 months in the PODCI Transfer and Basic Mobility Scale and PODCI Sports and Physical Functioning Scale, in which patients attained normal ranges by 6 months (FIG. 12A and FIG. 12B, respectively). The decrease in physical disability as assessed by the average PODCI scores was sustained throughout 5 years of treatment with asfotase alfa in both PODCI Transfer and Basic Mobility Scale and PODCI Sports and Physical Functioning Scales. In summary, decreased pain of the HPP children treated with asfotase alfa was apparent from CHAQ and PODCI discomfort median normative scores by 6 months, which was sustained throughout 5 years of treatment with asfotase alfa (FIGS. 13A and 13B, respectively). Thus, reduced pain and increased ability to perform ADL were sustained in this cohort of children with HPP throughout 5 years of treatment with asfotase alfa.

Example 8. Tolerability of HPP Children to Long-Term Treatment with Asfotase Alfa

Generally, treatment with asfotase alfa was well-tolerated in children having HPP, with most adverse events (AEs) considered mild or moderate. There were no deaths, serious AEs, or withdrawals due to AEs. One patient receiving asfotase alfa at a dosage regimen of 9 mg/kg/week subcutaneously underwent a dose reduction for a low plasma PLP level. Four patients had injection-associated reactions (10 events) during the first month of treatment. All were injection site reactions (ISRs), with the exception of nausea and peripheral edema in 1 patient, and all were mild. Each patient experienced at least 1 treatment-emergent AE; 86% mild and 14% moderate in severity. Most AEs (53%) were considered by the investigators to be unrelated to the treatment. When ISRs were excluded, 2% of AEs were deemed treatment related. The most common AEs were mild injection site reactions (Table 9). For example, erythematous reaction, purple discoloration at 5 years, and abdominal lipohypertrophy at 4.5 years were noted in individual patients.

TABLE 9 Occurrence of injection site reactions in patients treated with asfotase alfa for 5 years. Asfotase alfa, n = 13 Injection site reactions in ≥3 patients Events, n Patients, n (%) Injection site reactions 250 12 (92%) Erythema 71 11 (85%) Hypertrophy 26 8 (62) Pruritus 23 7 (54) Pain 18 6 (46) Atrophy 13 5 (38) Discoloration 17 5 (38) Swelling 12 3 (23) Induration 1 1 (8) Nodule 1 1 (8) Papule 1 1 (8) Urticaria 1 1 (8)

In summary, a total of 250 injection site reactions occurred in 12 of the 13 patients (92% of total patients). In particular, 71 instances of erythema occurred in 11 patients (85% of total patients); 26 instances of hypertrophy occurred in 8 patients (62% of total patients); 23 instances of pruritus occurred in 7 patients (54% of total patients); 18 instances of pain occurred in 6 patients (46% of total patients); 13 instances of atrophy occurred in 5 (38% of total patients); 17 instances of discoloration occurred in 5 patients (38% of total patients); 12 instances of swelling occurred in 3 patients (23% of total patients); 1 instance of induration occurred in 1 patient (8% of total patients); 1 instance of a nodule occurred in 1 patient (8% of total patients); 1 instance of a papule occurred in 1 patient (8% of total patients); and 1 instance of a urticaria occurred in 1 patient (8% of total patients).

Surveillance of radiographs of the HPP children treated with asfotase alfa showed no evidence of ectopic calcification. On ophthalmoscopy, the retinal examinations were consistently unremarkable. Notably, 46% (6 of 13) of the patients showed small refractile deposits in the conjunctiva or cornea presumed to contain calcium. For 5 HPP patients, these were considered possibly or probably related to the treatment. No deposit was deemed clinically apparent or significant. No children developed nephrocalcinosis, although after approximately 2 years of treatment, renal sonography revealed “focal echogenicity” consistent with a small renal stone present for 6 months in 1 kidney of 1 child.

All 12 treated HPP children showed anti-asfotase alfa antibodies. A total of 5 of the 6 patients who began treatment in the 3 mg/kg dose group were antibody positive by Week 6, and all were antibody positive at Week 12 of treatment. In those starting in the 2 mg/kg dose group, 1 patient was antibody positive by Week 6, and 5 patients were positive by Week 48. In 5 of the 12 patients, the antibodies were neutralizing in vitro at 1 or more time points, but with no apparent compromise of therapeutic efficacy.

Example 9. Study Design of HPP Infants Treated with Asfotase Alfa

Infants aged ≤3 years with onset of HPP symptoms prior to 6 months of age were selected for treatment with asfotase alfa for a time period of at least 5 years (see Table 10).

TABLE 10 Characteristics of HPP infants treated with asfotase alfa for 5 years. Characteristic asfotase alfa (N = 11) Age (months) 6.8 (0.7, 36.4) Age at onset of HPP (months) 1.0 (0.0, 5.8) Female, n (%) 7 (64) White, n (%) 10 (91) Failure to thrive, n (%) 10 (91) Length/height, Z-score −3.7 (−9.2, −0.7) Weight, Z-score −3.8 (−5.4, −0.5) BSID-III Gross motor 1 (1, 9) Fine motor 5 (1, 13)

Inclusion criteria included low alkaline phosphatase (ALP) activity (total serum ALP ≥3 SDs below the age-adjusted mean of healthy patients); high pyridoxal 5′-phosphate (PLP) plasma concentrations (plasma PLP ≥4 times upper limit of healthy patients); radiographic evidence of skeletal deformities (i.e. flared and frayed metaphyses, severe generalized osteopenia, and/or widened growth plates); rachitic chest deformity; vitamin B6—responsive seizures; failure to thrive; a history of non-traumatic postnatal fracture or delayed fracture healing; history of elevated serum calcium; craniosynostosis; nephrocalcinosis; and/or respiratory compromise. Exclusion criteria included low serum calcium, phosphate, or 25-hydroxyvitamin D levels.

Following the initial phase study, 10 of the 11 patients proceeded with an extension phase study of asfotase alfa administration for at least 5 years (one patient withdrew due to adverse events) (FIG. 14). In the extension phase, 9 out of 10 patients completed the study (one patient died due to septic shock unrelated to asfotase alfa administration). Asfotase alfa was administered via subcutaneous administration to patients at a dosage of 1.0 mg/kg three times weekly. Additional dose adjustments could be made for changes in weight and/or for safety concerns or for lack of efficacy after 1 month (e.g., increasing the dosage to 2 mg/kg three times weekly) and again after 3 months (e.g., increasing the dosage to 3 mg/kg three times weekly).

Example 10. Radiographic Assessments of HPP Infants Treated with Asfotase Alfa

Radiographic assessments of HPP patients were performed during the course of treatment as previously described (see Whyte et al. (N Engl J Med 366(10): 904-913, 2012)). The Radiographic Global Impression of Change (RGI-C) assessment scale was utilized during treatment of HPP patients with asfotase alfa. Paired pre-treatment and on-treatment radiographs of bilateral wrists, knees, and, when available, chests, were evaluated for changes in the skeletal manifestations of HPP (−3=severe worsening; 0=no change; +3=near/complete healing; FIG. 15A).

Evaluations were performed by three independent radiologists blinded to the post-baseline time point. The average RGI-C score was determined as the mean of the three RGI-C scores. A patient with a score of ≥+2 was considered a “responder” for that time point. RGI-C scores of the HPP patients demonstrated significant skeletal improvement by 6 months (primary endpoint), which was sustained throughout 5 years for the 9 remaining patients (FIG. 15B). For the primary endpoint analysis of RGI-C, a Wilcoxon signed-rank test with a two-sided alpha of 0.05 tested whether the median RGI-C at Week 24 differed from 0. If the P-value was <0.05 and the median RGI-C score was positive, significant positive change from baseline is shown. Missing RGI-C scores were imputed with last observation carried forward; patients with no post-baseline data were considered non-responders. Additionally, this responder analysis was repeated using the last assessment of RGI-C scores. From year 1, the majority of patients achieved RGI-C scores ≥2, corresponding to substantial healing of rickets.

Example 11. Growth Assessments of HPP Infants Treated with Asfotase Alfa

Length and height of HPP patients at each time point were measured, and BMI was determined from the length and height measurements. The CDC percentiles were generated using the 2000 National Center for Health Statistics Growth Chart Equation, Length Z-scores improved from baseline to 5 years of treatment, with scores reflecting catch-up growth relative to healthy peers (male patients shown in FIG. 16A; female patients shown in FIG. 16B). Likewise, weight Z-scores improved from baseline to 5 years of treatment, with scores reflecting catch-up growth relative to healthy peers (male patients shown in FIG. 17A; female patients shown in FIG. 17B). The median BMI and Z-scores (length and width) also exhibited improvement from baseline to 5 years of treatment (FIG. 18). Statistical significance was defined as P≤0.05.

Example 12. Motor Development of HPP Infants Treated with Asfotase Alfa

Motor development of HPP patients treated with asfotase alfa was determined using the Infant and Toddler Development, 3rd Edition (BSID-III) scaled score for patients ≤42 months of age and using the Locomotion Subtest of the Peabody Developmental Motor Scales, 2nd Edition (PDMS-2) standard score for patients 43 to 71 months of age. Detailed protocols are described in BSID-III, Bayley Scales of Infant and Toddler Development, 3rd Edition and in Peabody Developmental Motor Scales, 2nd edition.

BSID-III scaled score and PDMS-2 standard score both reflect the patient's performance relative to healthy, same-aged peers; the healthy age-matched mean for both scores was 10 (standard deviation of 3). At baseline or first assessment (FA), BSID-III median scaled scores indicated functional delay in all 11 patients (FIG. 19). BSID-III median scaled scores improved by 1 year of treatment, with continued improvement throughout asfotase alfa administration. The normal range of BSID-III scores was defined as described in Connolly et al. (Pediatr Phys Ther. 24(4):345-52, 2012; incorporated herein by reference). Five patients transitioned to PDMS-2 measurements by 5 years, the median scoring near the normal range for the PDMS-2 locomotion subtest (FIG. 20). Improvements in the PDMS-2 scaled score were noted through 5 years of therapy (median baseline score (min, max): 5 (1, 13). n=11; median score at year 2: 9.5 (7, 11), n=6; Year 3: 7 (7, 13), n=3). Calculation of the normal BSID-III range is described in Folio et al. Peabody developmental motor scales-2 (2nd ed.). Austin, Tex.; incorporated herein by reference. Substantial improvements in skeletal manifestations and motor function continued during 5 years of treatment for life-threatening perinatal and infantile HPP with asfotase alfa.

Example 13. Tolerability to Long-Term Treatment with Asfotase Alfa

Generally, treatment with asfotase alfa was well-tolerated in infants having HPP, with most adverse events (AEs) considered mild or moderate. The most common AEs of patients are described in Table 11. AH adverse events (AEs) were coded using the Medical Dictionary for Regulatory Activities (v. 13.0 or higher).

TABLE 11 Adverse events (AEs) of infants having HPP treated with asfotase alfa for 5 years. Preferred term Events, n Patients, n (%) Upper respiratory tract infection 67 7 (64%) Pyrexia 20 7 (64%) Pneumonia 14 7 (64%) Craniosynostosis 12 7 (64%) Otitis media 19 6 (55%) Vomiting 13 6 (55%) Constipation 7 6 (55%) Injection site erythema 29 5 (46%) Hemoglobin decreased 9 4 (36%) Tooth loss 8 4 (36%) Diarrhea 6 4 (36%) Nasopharyngitis 6 4 (36%) Rash 5 4 (36%) Dental caries 4 4 (36%) Irritability 4 4 (36%)

Injection associated reactions were reported by a total of 7 patients. Examples of common injection site reactions are shown in Table 12. In total, 73 injection site reactions occurred in 7 HPP patients (64% of total HPP patients) treated with asfotase alfa for 5 years. In particular, 28 instances of erythema occurred in 4 patients (37% of total HPP patients); 8 instances of hypertrophy occurred in 2 patients (18% of total HPP patients); 5 instances of both hematoma and swelling occurred in 2 patients (18% of total HPP patients); and 3 instances of nodule and warmth at the injection site occurred in 2 patients (18% of total HPP patients) throughout treatment with asfotase alfa for 5 years.

TABLE 12 Injection site reactions of infants having HPP treated with asfotase alfa for 5 years. Preferred term Events, n Patients, n (%) Injection Site Reactions 73 7 (64%) Erythema 28 4 (37%) Hypertrophy 8 2 (18%) Haematoma 5 2 (18%) Swelling 5 2 (18%) Nodule 3 2 (18%) Warmth 3 2 (18%)

Other Embodiments

All publications, patents, and patent applications mentioned in the above specification are hereby incorporated by reference to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety. Various modifications and variations of the described methods, pharmaceutical compositions, and kits of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the claimed invention. Although the disclosure has been described in connection with specific embodiments, it will be understood that it is capable of further modifications and that the invention as claimed should not be unduly limited to such specific embodiments.

Claims

1. A method of treating hypophosphatasia (HPP) in a child of about 5 to about 12 years of age having an average Bruininks-Oseretsky Test of Motor Proficiency 2nd Edition (BOT-2) strength score of less than about 10, wherein the method comprises administering a soluble alkaline phosphatase (sALP) to the child at a dosage providing about 6 mg/kg/week of the sALP, wherein the sALP comprises an amino acid sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 1, and wherein administration of the sALP results in an average increase in the BOT-2 strength score to about 10 up to about 20 after a period of at least four years.

2. The method of claim 1, wherein the average BOT-2 strength score of the child is determined relative to a BOT-2 strength score of a child without HPP.

3. The method of claim 2, wherein the child having an average BOT-2 strength score of less than about 10 exhibits physical impairments relative to the child without HPP.

4. The method of any one of claims 1 to 3, wherein the child exhibits a decrease in physical impairments after administration of the sALP.

5. The method of claim 4, wherein the decrease in physical impairments is sustained throughout administration of the sALP to the child.

6. The method of any one of claims 1 to 5, wherein the average BOT-2 strength score of the child increases to about 12 to about 16.

7. The method of any one of claims 1 to 6, wherein the average BOT-2 strength score of the child is determined from measurements selected from the group consisting of sit-ups, V-ups, standing long jump, wall sit, and push-ups.

8. A method of treating HPP in a child of about 5 to about 12 years of age having an average BOT-2 running speed and agility score of less than 5, wherein the method comprises administering a sALP to the child at a dosage providing about 6 mg/kg/week of the sALP, wherein the sALP comprises an amino acid sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 1, and wherein administration of the sALP results in an average increase in the BOT-2 running speed and agility score to about 5 up to about 20 after a period of at least four years.

9. The method of claim 8, wherein the average BOT-2 running speed and agility score of the child is determined relative to a BOT-2 running speed and agility score of a child without HPP.

10. The method of claim 9, wherein the child having an average BOT-2 running speed and agility score of less than 5 exhibits physical impairments relative to the child without HPP.

11. The method of any one of claims 8 to 10, wherein the child exhibits a decrease in physical impairments after administration of the sALP.

12. The method of claim 11, wherein the decrease in physical impairments is sustained throughout administration of the sALP to the child.

13. The method of any one of claims 8 to 12, wherein the average BOT-2 running speed and agility score of the child increases to about 9 to about 13.

14. The method of any one of claims 8 to 13, wherein the average BOT-2 running speed and agility score of the child is determined from measurements selected from the group consisting of stepping over a balance beam, shuttle run, two-legged side hop, and one-legged side hop.

15. The method of any one of claims 1 to 14, wherein said method further comprises performing a BOT-2 test.

16. A method of treating HPP in a child of about 5 to about 12 years of age having an average Childhood Health Assessment Questionnaire (CHAQ) disability index and/or discomfort score of greater than about 0.8, wherein the method comprises administering a sALP to the child at a dosage providing about 6 mg/kg/week of the sALP, wherein the sALP comprises an amino acid sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 1, and wherein administration of the sALP results in an average decrease in the CHAQ disability index score and/or discomfort score to about 0 up to equal to or less than about 0.5 after a period of at least four years.

17. The method of claim 16, wherein the average CHAQ disability index and/or discomfort score of the child is determined relative to a CHAQ index score of a child without HPP.

18. The method of claim 17, wherein the child having an average CHAQ disability index and/or discomfort score of greater than 0.5 exhibits disability in activities of daily living (ADL) and pain, relative to the child without HPP.

19. The method of any one of claims 16 to 18, wherein the child exhibits an increase in ADL after administration of the sALP.

20. The method of claim 19, wherein the increase in ADL is sustained throughout administration of the sALP to the child.

21. The method of any one of claims 16 to 20, wherein the child exhibits a decrease in pain after administration of the sALP.

22. The method of claim 21, wherein the decrease in pain is sustained throughout administration of the sALP to the child.

23. The method of any one of claims 16 to 22, wherein the average CHAQ disability index score and/or CHAQ discomfort score of the child decreases to about 0 up to equal to or less than about 0.25.

24. The method of any one of claims 16 to 23, wherein said method further comprises performing a CHAQ test.

25. A method of treating HPP in a child of about 5 to about 12 years of age having an average Pediatric Outcomes Data Collection Instrument (PODCI) transfer and mobility, sports and physical functioning, and/or pain score of less than about 40, wherein the method comprises administering a sALP to the child at a dosage providing about 6 mg/kg/week of the sALP, wherein the sALP comprises an amino acid sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 1, and wherein administration of the sALP results in an average increase in the PODCI score to about 40 up to about 50 after a period of at least four years.

26. The method of claim 25, wherein the average PODCI transfer and mobility, sports and physical functioning, and/or pain score of the child is determined relative to a PODCI score of a child without HPP.

27. The method of claim 26, wherein the child having an average PODCI transfer and mobility, sports and physical functioning, and/or pain score of less than 40 exhibits disability in ADL and pain relative to the child without HPP.

28. The method of any one of claims 25 to 27, wherein the child exhibits an increase in ADL after administration of the sALP.

29. The method of claim 28, wherein the increase in ADL is sustained throughout administration of the sALP to the child.

30. The method of any one of claims 25 to 29, wherein the child exhibits a decrease in pain after administration of the sALP.

31. The method of claim 30, wherein the decrease in pain is sustained throughout administration of the sALP to the child.

32. The method of any one of claims 25 to 31, wherein said method further comprises performing a PODCI test.

33. A method of treating HPP in a child of about 5 to about 12 years of age, wherein the method comprises administering a sALP to the child at a dosage providing about 6 mg/kg/week of the sALP, wherein the sALP comprises an amino acid sequence having at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 1, and wherein administration of the sALP for a treatment period of at least one year results in an average increase in a Six Minute Walk Test (6MWT) distance of the child relative to the 6MWT distance of the child prior to administration of the sALP.

34. The method of claim 33, wherein the average increase in the 6MWT distance is about 20 meters or greater.

35. The method of claim 34, wherein the average increase in the 6MWT distance is about 20 meters, about 25 meters, about 30 meters, about 35 meters, about 40 meters, about 45 meters, about 50 meters, about 55 meters, about 60 meters, about 65 meters, about 70 meters, about 75 meters, about 80 meters, about 85 meters, about 90 meters, about 95 meters, about 100 meters, or greater.

36. The method of any one of claims 33 to 35, wherein the average increase in the 6MWT value of the patient is sustained during a treatment period of at least two years, at least three years, at least four years, or longer.

37. The method of any one of claims 33 to 36, wherein the child exhibits an improvement in rickets as determined by the Rickets Severity Scale (RSS) after administration of the sALP.

38. The method of any one of claims 33 to 37, wherein the child exhibits an increase in ADL after administration of the sALP.

39. The method of claim 38, wherein the increase in ADL is determined by a CHAQ disability index score, PODCI global function scale score, PODCI transfer and mobility scale score, or PODCI sports and physical functioning scale score.

40. The method of any one of claims 33 to 39, wherein the 6MWT distance of the child is determined relative to the 6MWT distance of a healthy child or an untreated child having HPP.

41. The method of any one of claims 33 to 40, wherein said method further comprises performing a 6MWT.

42. The method of any one of claims 1 to 41, wherein the child of about 5 to about 12 years of age exhibited an average Bayley Scales of Infant and Toddler Development, 3rd Edition (BSID-III) scaled score of less than about 2 at about 3 years of age or less than 3 years of age, wherein administration of the sALP results in an average increase in the BSID-III scaled score to greater than about 5 after a period of at least four years.

43. The method of claim 42, wherein the average BSID-III scaled score of the child is determined relative to an average BSID-III scaled score of a child without HPP.

44. The method of claim 43, wherein the child having an average BSID-III scaled score of less than 2 exhibits delayed motor development relative to a child without HPP.

45. The method of any one of claims 42 to 44, wherein the child exhibits an increase in motor development after administration of the sALP.

46. The method of claim 45, wherein the increase in motor development is sustained throughout administration of the sALP to the child.

47. The method of claim 46, wherein the average BSID-III scaled score increases to about 5, to about 6, or to about 7.

48. The method of any one of claims 42 to 47, wherein the average BSID-III scaled score of the child is determined from measurements selected from the group consisting of prehension, perceptual-motor integration, motor planning and speed, visual tracking, reaching, object grasping, object manipulation, functional hand skills, responses to tactile information, movement of the limbs and torso, static positioning, dynamic movement, balance, and motor planning.

49. The method of any one of claims 42 to 48, wherein said method further comprises performing a BSID-III test.

50. The method of any one of claims 1 to 49, wherein the child of about 5 to about 12 years of age exhibited an average Peabody Developmental Motor Scales, 2nd Edition (PDMS-2), standard score of about 5 at about 5 years of age or less than 5 years of age, and wherein administration of the sALP results in an average increase in the PDMS-2 standard score to about 7 after a period of at least four years.

51. The method of claim 50, wherein the average PDMS-2 standard score of the child is determined relative to a PDMS-2 standard score of a child without HPP.

52. The method of claim 51, wherein the child having an average PDMS-2 standard score of about 5 exhibits delayed motor development relative to the child without HPP.

53. The method of any one of claims 50 to 52, wherein the child exhibits an increase in motor development after administration of the sALP.

54. The method of claim 53, wherein the increase in motor development is sustained throughout administration of the sALP to the child.

55. The method of any one of claims 50 to 54, wherein said method further comprises performing a PDMS-2 test.

56. The method of any one of claims 50 to 55, wherein the average PDMS-2 standard score of the child is determined from measurements selected from the group consisting of crawling, walking, running, hopping, jumping forward, reflexes, balance, object manipulation, grasping, and visual-motor integration.

57. The method of any one of claims 1 to 56, wherein the sALP is formulated for daily or weekly administration.

58. The method of any one of claims 1 to 57, wherein the sALP is formulated for administration twice a week, three times a week, four times a week, five times a week, six times a week, or seven times a week.

59. The method of any one of claims 1 to 58, wherein the sALP is formulated at a dosage of 2 mg/kg for administration three times a week.

60. The method of any one of claims 1 to 59, wherein the sALP is formulated at a dosage of 1 mg/kg for administration six times a week.

61. The method of any one of claims 1 to 60, wherein the sALP is formulated at a dosage of 3 mg/kg for administration three times a week.

62. The method of any one of claims 1 to 61, wherein the sALP is administered for at least five years, at least six years, at least seven years, at least eight years, at least nine years, at least ten years, or more than ten years.

63. The method of any one of claims 1 to 62, wherein the sALP comprises the amino acid sequence of SEQ ID NO: 1.

64. The method of any one of claims 1 to 63, wherein the sALP consists of the amino acid sequence of SEQ ID NO: 1.

65. The method of any one of claims 1 to 64, wherein the method further comprises determining sALP activity of a child.

66. The method of claim 65, wherein the determination of sALP activity comprises measuring at least one of phosphoethanolamine (PEA), inorganic pyrophosphate (PPi), and pyridoxal 5′-phosphate (PLP) in at least one of a serum sample and blood sample from the child.

67. The method of any one of claims 1 to 66, wherein the sALP is administered in an amount that is therapeutically effective to treat at least one symptom of HPP.

68. The method of claim 67, wherein the at least one symptom of HPP comprises rickets, premature loss of deciduous teeth, incomplete bone mineralization, elevated blood and/or urine levels of inorganic pyrophosphate (PR), elevated blood and/or urine levels of phosphoethanolamine (PEA), elevated blood and/or urine levels of pyridoxal 5′-phosphate (PLP), hypomineralization, rachitic ribs, hypercalciuria, short stature, skeletal deformity, waddling gait, bone pain, bone fracture, HPP-related seizure, inadequate weight gain, and calcium pyrophosphate dihydrate crystal deposition.

69. The method of any one of claims 1 to 68, wherein the sALP is administered at an initial dosage of about 3 mg/kg/week and then increased to a dosage of about 6 mg/kg/week or more.

70. The method of claim 69, wherein the dosage is increased to about 6 mg/kg/week or more after about three months to about nine months.

71. The method of claim 69 or 70, wherein the dosage is increased to about 9 mg/kg/week.

72. The method of any one of claims 1 to 71, wherein symptoms of HPP are exhibited at birth.

73. The method of any one of claims 1 to 72, wherein symptoms of HPP are not exhibited at birth.

74. The method of any one of claims 1 to 73, wherein symptoms of HPP are exhibited at five years of age or older.

75. The method of any one of claims 1 to 74, wherein the child exhibits tolerability to administration of the sALP.

76. The method of claim 75, wherein the tolerability comprises a lack of or decreased incidence of at least one adverse event selected from the group consisting of injection site erythema, decrease in hemoglobin, pyrexia, pneumonia, upper respiratory tract infection, craniosynostosis, otitis media, vomiting, constipation, diarrhea, tooth loss, nasopharyngitis, rash, dental carries, and irritability.

77. The method of any one of claims 1 to 76, wherein the child does not exhibit at least one of a serum calcium level and a phosphorus level below the age-adjusted normal range.

78. The method of any one of claims 1 to 77, wherein the child does not exhibit symptoms of a treatable form of rickets.

79. The method of any one of claims 1 to 78, wherein the child has not previously received treatment with a bisphosphonate.

80. The method of any one of claims 1 to 79, wherein the method further comprises performing radiographs of the child to determine an average Radiographic Global Impression of Change (RGI-C) score.

81. The method of claim 80, wherein the child exhibits an average RG1-C score of less than 2 prior to administration of the sALP.

82. The method of claim 80 or 81, wherein the average RGI-C score of the child is determined relative to an average RGI-C score of a child without HPP.

83. The method of any one of claims 80 to 82, wherein the child exhibits increased bone density after administration of the sALP.

84. The method of claim 83, wherein the increase in bone density is sustained throughout administration of the sALP.

85. The method of claim 84, wherein the child exhibits an average RGI-C score of greater than 2.

86. The method of any one of claims 1 to 85, wherein the method further comprises determining weight and/or length of the child.

87. The method of claim 86, wherein a Z-score is determined from the weight and/or length of the child.

88. The method of claim 87, wherein the child exhibits an improved Z-score for weight and/or length after administration of the sALP.

89. The method of any one of claims 1 to 88, wherein the sALP is formulated in a pharmaceutical composition, with at least one pharmaceutically acceptable carrier.

90. The method of claim 89, wherein the at least one pharmaceutically acceptable carrier is saline.

91. The method of claim 90, wherein the at least one pharmaceutically acceptable carrier comprises sodium chloride and sodium phosphate.

92. The method of claim 91, wherein the at least one pharmaceutically acceptable carrier comprises 150 mM sodium chloride and 25 mM sodium phosphate.

93. The method of any one of claims 89 to 92, wherein the pharmaceutical composition is formulated for at least one of subcutaneous, intramuscular, intravenous, oral, nasal, sublingual, intrathecal, and intradermal administration.

94. The method of claim 93, wherein the pharmaceutical composition is formulation for subcutaneous administration.

95. The method of any one of claims 1 to 94, wherein the sALP is physiologically active toward PEA, PPi, and PLP.

96. The method of any one of claims 1 to 95, wherein the sALP is catalytically competent to improve skeletal mineralization in bone.

97. The method of any one of claims 1 to 96, wherein the sALP is the soluble extracellular domain of an alkaline phosphatase.

98. The method of any one of claims 1 to 97, wherein the child has not previously been administered the sALP.

Patent History
Publication number: 20190099473
Type: Application
Filed: Sep 1, 2016
Publication Date: Apr 4, 2019
Patent Grant number: 11065306
Inventors: Kenji FUJITA (Millburn, NJ), Dawn PHILLIPS (Chapel Hill, NC)
Application Number: 16/083,186
Classifications
International Classification: A61K 38/46 (20060101); C12N 9/16 (20060101); A61P 19/08 (20060101);